US20170199184A1 - Capture, purification, and release of biological substances using a surface coating - Google Patents

Capture, purification, and release of biological substances using a surface coating Download PDF

Info

Publication number
US20170199184A1
US20170199184A1 US15/378,938 US201615378938A US2017199184A1 US 20170199184 A1 US20170199184 A1 US 20170199184A1 US 201615378938 A US201615378938 A US 201615378938A US 2017199184 A1 US2017199184 A1 US 2017199184A1
Authority
US
United States
Prior art keywords
microfluidic chip
composition
surface coating
cells
nonfouling
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/378,938
Inventor
Ying-Chih Chang
Han-Chung Wu
Po-Yuan Tseng
Jen-Chia Wu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Academia Sinica
Original Assignee
Academia Sinica
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Academia Sinica filed Critical Academia Sinica
Priority to US15/378,938 priority Critical patent/US20170199184A1/en
Publication of US20170199184A1 publication Critical patent/US20170199184A1/en
Priority to US16/832,396 priority patent/US11674958B2/en
Priority to US18/308,531 priority patent/US20230366879A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54393Improving reaction conditions or stability, e.g. by coating or irradiation of surface, by reduction of non-specific binding, by promotion of specific binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K17/00Carrier-bound or immobilised peptides; Preparation thereof
    • C07K17/14Peptides being immobilised on, or in, an inorganic carrier
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/40Concentrating samples
    • G01N1/405Concentrating samples by adsorption or absorption
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • G01N33/54386Analytical elements
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells

Definitions

  • Table 1 is the amino acid sequence of EpAb4-1 antibody.
  • the shedding of cells into the circulation is an intrinsic property of the malignant tumor, and this feature provides important information with regard to the diagnosis, staging, treatment response and survival of cancer patients.
  • Pantel et al found the number of circulating tumor cells (CTCs) in the blood is correlated with the aggressiveness of the cancer as well as the efficacy of the therapy. (Pantel, K. et. al., “Detection, clinical relevance and specific biological properties of disseminating tumor cells”, Nat Rev Cancer, 2008, 8(5):329-40).
  • CFCs as few as one per 109 blood cells in patients with metastatic cancer, are rare cells. This makes the detection and isolation of CTCs technically challenging (sec Kahnet al. Breast Cancer Res Treat 2004, 86:237-47). An enrichment process is therefore necessary to effectively detect and isolate CTCs.
  • An example of such enrichment process is the use of a highly overexpressed cell surface biomarker with high specificity and sensitivity for CTCs, such as the epithelial cell adhesion molecule (EpCAM).
  • EpCAM epithelial cell adhesion molecule
  • the Cellsearch SystemTM (Veridex), the only FDA-approved platform for CTC detection, utilizes anti-EpCAM antibody-coated magnetic nanoparticles to capture and enrich CTCs, followed by cytokeratin immunostaining.
  • the AdnaTest AdnaGen AG, Germany
  • another commercially available system for CTC detection adopts similar immunomagnetic approach by using anti-EpCAM and Mucin 1 (MUC1) conjugated magnetic beads.
  • CTC chips based on anti-EpCAM antibody-coated microfluidics chip were developed for CTC detection and enrichment (Nagrath et al, Nature 2007, 450:1235-9).
  • the disadvantage of the above techniques is the low detection rate of pure CTCs, due to the non-specific binding of blood cells with anti-EpCAM antibody.
  • Kaladhar et al. observed a significant fewer circulating blood cells (e.g. platelets, leukocytes, and erythrocytes) binding onto the solid substrate modified with supported monolayer of various lipid compositions containing phosphatidyl-choline, cholesterol, and glycolipid (Kaladhar et al, Langmuir 2004, 20: 11115-22 and Kaladhar et al, J Biomed Mater Res A 2006, 79A:23-35).
  • circulating blood cells e.g. platelets, leukocytes, and erythrocytes
  • the present invention is directed to a surface coating to capture a circulating rare cell (CRC).
  • CRC circulating rare cell
  • This surface coating increases the capture efficiency of a CRC, such as CTC, circulating stem cells (e.g. tumor stem cell and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like and reduces the binding of non-specific cells or protein adsorption.
  • CRC circulating rare cell
  • the surface coating comprises 1) a nonfouling composition that reduces the binding of nonspecific blood cells and adsorption of other blood components, such as protein; and 2) a bioactive composition that captures a CRC.
  • the surface coating further comprises a linker composition that attaches to the nonfouling composition and the bioactive composition, as illustrated in FIG. 1A .
  • the present invention is directed to a surface coating to capture and release a biological substance.
  • This surface coating increases the capture efficiency of a biological substance, such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like and enhances the removal or release of the non-specific cells or protein from the surface coating.
  • the surface coating comprises 1) a releasable composition for releasing or removing nonspecific blood cells and other blood components, such as protein, from the surface coating; and 2) a bioactive composition that captures a biological substance.
  • the surface coating further comprises a linker composition that attaches to the releasable composition and the bioactive composition.
  • the present invention is also directed to a microfluidic device, with specific microstructure designs to create a disturbed flow of blood, body fluid or biological samples to increase the capture rate of the biological substance.
  • the present invention is also directed to a method of manufacturing the surface coating, comprising a) forming the nonfouling or the releasable composition; and b) attaching the the linker composition with the nonfouling/releasable composition from step a) and the bioactive composition, or c) attaching the nonfouling/releasable composition from step a) with the bioactive composition.
  • the present invention is also directed to methods to capture and release the biological substance from the surface coating.
  • the biological substance on the surface coating can be purified by removing the non-specific cells or protein.
  • the captured biological substance can be released by air bubbles, ultraviolet irradiation and the like.
  • the present invention is also directed to uses of a biotinylated anti-EpCam antibody, EpAb4-1 antibody, to capture a CTC.
  • FIG. 1A illustrates schematically an embodiment of the surface coating comprising a nonfouling composition, a linker composition and a bioactive composition.
  • FIG. 1B illustrates schematically the binding of a circulating tumor cell with the surface coating from FIG. 1A
  • FIG. 2A to FIG. 2F illustrate the chemical structures of examples of nonfouling materials.
  • FIG. 3 illustrates the chemical reactions of conjugation between the functional groups on the nonfouling composition and the bioactive composition.
  • FIG. 4A illustrates schematically the attachment of the surface coating and solid substrate without a surface linker.
  • FIG. 4B and FIG. 4C illustrate schematically a linker composition with a cleavable functional group.
  • FIG. 4D illustrates schematically the attachment of the surface coating and the solid substrate using a surface linker.
  • FIG. 5A and FIG. 5B illustrates schematically the formation of the surface coating on a solid substrate.
  • FIGS. 6A and 6B illustrate schematically the components of a microfluidic chip.
  • FIG. 6C illustrates schematically the microfluidic chip assembly to capture CTCs from a biological sample.
  • FIG. 7A to FIG. 7H illustrate schematically the designs of the microstructures the solid substrate.
  • FIGS. 7I and 7J illustrate the capture efficiency of various microstructure designs in DMEM solution and blood respectively.
  • FIG. 8 illustrates the shear stresses of a buffer solution to release the non-specific cells and purify the captured biological substance.
  • FIG. 9 illustrates schematically the release of biological substance by the air bubble method.
  • FIG. 10A illustrates schematically lie surface coating with a cleavable linker composition on a solid substrate.
  • FIG. 10B illustrates schematics the release of the biologic substance from the surface coating in FIG. 10A .
  • FIG. 11 illustrates QCM-D response of the surface coating construction.
  • FIG. 12 illustrates the QCM-D response of the addition of bovine serum albumin the surface coating.
  • FIG. 13 are the photographs of the non-specific cells (top images) and the CTCs (bottom images) on the surface coating before and after the buffer rinse.
  • FIG. 14A illustrates the capture efficiency and non-specific blood cell binding of various surface coatings.
  • FIG. 14B are photo images which illustrate the non-specific blood cell binding of various surface coatings before and after the buffer rinse.
  • FIG. 15A to FIG. 15C are the photographs of the non-specific cells and the biological substance on the surface coating before and after the buffer rinse purification.
  • FIG. 16 illustrates the various shear stress and flushing time for the removal of HCT116 and NTH-3T3 cell populations from the surface coating.
  • FIG. 17 are the photographs of the CTCs released by the air bubbles.
  • FIG. 18 illustrates the cell cultures of the released CTCs on day 1day 10 and day 14.
  • FIG. 19 illustrates schematically a CTC filtration device.
  • FIG. 20 illustrates the CTC binding specificity of biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody, biotinylated EpCam antibody and IgG antibody,
  • the present invention is directed to a surface coating to effectively capture a circulating rare cell (CRC), such as CTC, circulating stem cells (e.g. tumor stem cell and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like.
  • CTC circulating rare cell
  • stem cells e.g. tumor stem cell and bone marrow stem cells
  • fetal cells fetal cells
  • bacteria fetal cells
  • virus epithelial cells
  • endothelial cells or the like.
  • the surface coating for the capture of a CRC comprises 1) a nonfouling composition that prevents the binding of non-specific cells and adsorption of other blood components, such as protein; and 2) a bioactive composition that captures the circulating rare cells.
  • the nonfouling composition and the bioactive composition are joined by discrete functional groups or moieties present in the nonfouling and bioactive compositions.
  • a linkage between the two compositions is formed by an interaction comprising electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, complementary DNA binding, magnetic force, or combinations thereof.
  • complementary DNA fragments are used for binding the nonfouling composition and the bioactive composition.
  • the fragments are attached to each of the compositions and can be partially or completely complementary over their lengths.
  • a suitable length of DNA will generally be at least 15, 20, 25, 35, 50, 100 or more bases in length.
  • An example of the DNA used in the present invention is an DNA tweezer. (See, B Yurke et al., A DNA-fuelled molecular machine made of DNA. Nature 2000, 406:605-608.)
  • the surface coating comprises 1) a nonfouling composition; 2) a bioactive composition; and 3) a linker composition, which joins the nonfouling composition to the bioactive composition. See FIG. 1A .
  • the present invention is also directed to a surface coating to effectively capture a biological substance, such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like, purify the biological substance on the surface of the surface coating by releasing or removing the non-specific cells and other serum components (e.g. protein) through a buffer rinse, and release the captured biological substance from the surface coating.
  • a biological substance such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like
  • serum components e.g. protein
  • the surface coating for the capture and purification of a biological substance comprises 1) a releasable composition for releasing nonspecific blood cells and other blood components, such as protein, through a buffer rinse; and 2) a bioactive composition that captures a biological substance.
  • the releasable composition and the bioactive composition are joined by discrete functional groups or moieties present in the releasable and bioactive compositions.
  • a linkage between the two compositions is formed by an interaction comprising electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, complementary DNA binding, magnetic force, or combinations thereof.
  • the surface coating further comprises a linker composition that attaches to the releasable composition and the bioactive composition.
  • the surface coating can be incorporated into the following configurations: cell cultural dishes, microfluidic channels, microfluidic chips, filtration filter, capillaries, tubes, beads, nanoparticies, or the like, with an inner diameter ranging from about 50 to about 1000 um.
  • nonfouling composition reduces the binding of non-specific cells and adsorption of the serum protein.
  • the “releasable” composition comprises a nonfouling composition which also acts as a “lubricating” surface such that only low flow shear stress is required to remove or release the non-specific cells or blood components from the surface coating, while the biological substance remains intact.
  • the nonfouling composition is selected from the group consisting of: a supported lipid layer such as liposomes, supported lipid bilayers (SLBs) or lipid multilayer, polypeptides, polyelectrolyte multilayers (PEMs), polyvinyl alcohol, polyethylene glycol (PEG) as illustrated in FIG. 2A , hydrogel polymers, extracellular matrix proteins, carbohydrate, polymer brushes, zwitterionic materials such as poly(carboxybetaine) (pCB)) as illustrated in FIG. 2D , poly(sulfobetaine) (pSB) as illustrated in FIG. 2E and pDMAEMA as illustrated in FIG. 2F , small organic compounds, and the combination of above materials forming a single or a multi-layer.
  • a supported lipid layer such as liposomes, supported lipid bilayers (SLBs) or lipid multilayer
  • polypeptides polyelectrolyte multilayers (PEMs)
  • PEG polyethylene glyco
  • the nonfouling composition comprises supported lipid bilayers (SLBs)
  • the SLBs typically comprise lipids such as, for example, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(cap biotinyl) (sodium salt) (b-PE) as illustrated in FIG. 2B and 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC).
  • lipids such as, for example, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(cap biotinyl) (sodium salt) (b-PE) as illustrated in FIG. 2B and 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC).
  • POPC 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine
  • the protein resistant property of a SLB can be explained by the presence of neutral and zwitterionic phosphatidylcholine headgroups in a wide pH range, as well as an aqueous thin film formed between the hydrophilic lipid head groups and the bulk solution (see, Johnson et al., Biophys J 1991, 59:289-94).
  • the nonfouling composition comprises PEG, preferably PEG with a molecular weight from about 100 to about 100,000 and exhibits a nonfouling property.
  • the nonfouling composition comprises polyelectrolyte multilayers (PEMS) or a polymer brush.
  • PEMs polyelectrolyte multilayers
  • suitable PEMs include, but are not limited to, poly-L-lysine/poly-L-glutamine acid (PLL/PLGA), poly-L-lysine/poly-L-aspartic acid or similar counter ionic polyelectrolytes.
  • the polymer brush comprises ([2-(acryloyloxy)ethyl] trimethyl ammonium chloride, TMA)/(2-carboxy ethyl acrylate, CAA) copolymer as illustrated in FIG. 2C .
  • TMA acryloyloxyethyl
  • CAA 2-carboxy ethyl acrylate copolymer as illustrated in FIG. 2C .
  • the nonfouling layer has a thickness from a few nanometers up to hundreds microns.
  • the nonfouling composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment, either directly to a functional group present in the bioactive composition, or directly to a functional group that is part of the linkage composition.
  • the functional groups of the nonfouling composition are selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the linker or bioactive composition.
  • the functional groups of the nonfouling composition which are first members of a binding pair, are selected from the group using specific binding recognition consisting of avidin, streptavidin DNA, RNA, ligand, receptor, antigen, antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present in either the linker or bioactive composition.
  • the linker composition joins the nonfouling/releasable composition and the bioactive composition and comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling/releasable composition and to a functional group that is part of the bioactive composition.
  • the linker composition comprises functional groups (prior to covalent attachment) selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the nonfouling or bioactive composition.
  • functional groups selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the nonfouling or bioactive composition.
  • the linker composition comprises functional groups (prior to non-covalent attachment) which are first members of a binding pair, selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen, antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present on the nonfouling/releasable composition or the bioactive composition.
  • the functional groups on the linker composition can also be a cleavable functional group, selected from: a photosensitive functional group cleavable by ultraviolet irradiation, an electrosensitive functional group cleavable by electro pulse mechanism, a magnetic material cleavable by the absence of the magnetic force, a polyelectrolyte material cleavable by breaking the electrostatic interaction, a DNA cleavable by hybridization, and the like.
  • the bioactive composition joins to either the linker composition or the nonfouling composition, and comprises a binding moiety selective for the detection of the biological substance or CRC.
  • the bioactive composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling layer or to a functional group that is part of the linker composition.
  • the functional groups of the bioactive composition are selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups which are selected to be reactive with functional groups present in either the nonfouling or linker composition.
  • the functional groups of the bioactive composition are selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen--antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present on the nonfouling/releasable composition or the linker composition.
  • the binding moiety of the bioactive composition has specific affinity with the biological substance through molecular recognition, chemical affinity, or geometrical/shape recognition.
  • the binding moiety for the detection of the biological substance include, but are not limited to: synthetic polymers, molecular imprinted polymers, extracellular matrix proteins, binding receptors, antibodies, DNA, RNA, antigens or any other surface markers which present high affinity to the biological substance.
  • a preferred antibody is the anti-EpCAM membrane protein antibody (commercially available from many sources, including R&D Systems, MN, USA), which provides high specificity for CTCs because EpCAM is frequently overexpressed in the lung, colorectal, breast, prostate, head and neck, and hepatic malignancies, but is absent from haematologic cells.
  • Anti-HER2 which has high specificity for CFCs but absent in haematologic cells.
  • the anti-EpCAM membrane protein antibody is EpAb4-1 antibody, comprising a heavy chain sequence with SEQ ID No:1 and a light chain sequence with SEQ ID NO: 2 shown in Table 1.
  • the bioactive composition can have a variety of thicknesses, selected so that it does not affect the function or the performance of the surface coating.
  • the conjugation linkers or catalysts for the nonfouling composition and the bioactive compositions are biotin/avidin or their derivatives.
  • the conjugation linkers or catalysts for the nonfouling composition and the bioactive composition are EDC/NHS.
  • the conjugation linkers or catalysts for the nonfouling composition and the bioactive compositions are sulfo-SMCC.
  • FIG. 3 schematically illustrates the chemical reactions of these embodiments.
  • the surface coating is attached to the solid substrate without a surface linker, as illustrated in FIG. 4A .
  • the nonfouling/releasable composition is attached to the solid substrate via one of the following interactions: covalent bonding (for PEG nonfouling composition), hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction (for SLB nonfouling/releasable composition), polar-polar interaction, complimentary DNA binding, magnetic force, or the like.
  • the surface coating is attached to the solid substrate with a surface linker, as illustrated in FIG. 4D .
  • the solid substrate used in the present invention include, but are not limited to: metals, plastics, glass, silicon wafers, hydroxylated poly(methyl methacrylate) (PMMA), and a combination thereof.
  • PMMA hydroxylated poly(methyl methacrylate)
  • the shape of the solid substrate include, but are not limited to: planar, circular and irregular shapes with micro, or nano-structures such as nanoparticles, nanowires, and a combination thereof.
  • the surface linker composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling/releasable composition and to a functional group that is part of the solid substrate.
  • Examples of the surface linker for binding the surface coating to a glass substrate include, but are not limited to, silane, aminopropyltriethoxy aminopropyltrimethoxy silane, silane-PEG-NH 2 , silane-PEG-N 3 (PEG molecular weight is about 1,000 to about 30,000 daltons) and silane-PEG biotin.
  • the surface linker comprises a cleavable functional group selected from: a photosensitive functional group cleavable by ultraviolet irradiation, an electrosensitive functional group cleavable by electro-pulse mechanism, an iron or magnetic material in which the absence of the magnetic force will release the nonfouling composition a polyelectrolyte material cleavable by breaking the electrostatic interaction, an DNA cleavable by hybridization, and the like.
  • the nonfouling composition comprises silane-functionalized PEG and the solid substrate is preferably selected from the group consisting of silicon, glass, hydroxylated poly(methyl methacrylate) (PMMA) aluminum oxide, TiO 2 and the like.
  • the nonfouling composition comprises thiol-functionalized compounds and the solid substrate is preferably selected from the group consisting of Au, Ag, Pt, and the like.
  • FIGS. 5A and 5B show the steps of forming the surface coating:
  • he surface coating without a linker composition can be formed by:
  • the microfluidic chip comprises a first solid substrate 1 (e.g. PMMA) and a second solid substrate 2 (e.g. glass), wherein the first and second solid substrates are adhered together using an adhesive means 3 or other means.
  • first solid substrate 1 e.g. PMMA
  • second solid substrate 2 e.g. glass
  • the surface of one or both solid substrates can be engraved with microstructures 4 .
  • the microstructures 4 are arranged in a linear fashion.
  • the microstructures 4 are arranged in herringbone fashion.
  • the shaded region on the adhesive 3 in FIG. 6B is carved out to accommodate the microstructures 4 on the surface of the solid substrate 1 .
  • a sealed channel 5 is created by adhering the first solid substrate 1 and the second solid substrate 2 together with an adhesive 3 .
  • the height of the channel 5 is determined by the thickness of the adhesive 3 .
  • the surface coating can be attached to one or both solid substrates.
  • the surface coating is attached to the solid substrate with a surface linker.
  • the surface coating is attached to the solid substrate via one of the following interactions: covalent bonding (for PEG nonfouling composition), hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction (for SLB nonfouling/releasable composition), polar-polar interaction, complimentary DNA binding, magnetic force, or the like.
  • the microstructures 4 on the solid substrate arc perpendicular to the flow direction and create a chaotic or disturbed flow of the blood, body fluid or biologic sample as it passes through the sealed channel 5 of the microfluidic chip.
  • the disturbed flow enhances the biological substance-surface coating contact.
  • FIG. 7A shows various designs of the microstructures 4 on the solid substrate.
  • the microstructures in Design F are arranged in a herringbone pattern whereas the microstructures in Designs A-E and H are arranged in a linear pattern.
  • the dimensions of the microstructures 4 are as follows: the length is about 50 mm fir O-D and G and about 120 mm for E-F, the height is about 30 ⁇ m, the width is about 1.5 mm for O and A, about 3.0 mm for B, and about 5..5 mm for C-G.
  • the height of the sealed channel 5 varies with the thickness of the adhesive 3 , preferably about 30-90 ⁇ m, more preferably about 60 ⁇ m.
  • FIG. 7B-7H show the details of Designs A-C in FIG. 7A .
  • Design C in FIG. 7H is the preferred pattern, with the following dimensions: the width of Microstructure (W) is about 150 ⁇ m, the length of microstructure (L) is about 1000 ⁇ m, the distance between two rows of microstructures (Sr) is about 250 ⁇ m, the distance between two adjacent microstructures (Sc) is about 350 ⁇ m, the height of the microstructure (D) is about 30 ⁇ m and the height of the sealed channel 5 (H) is about 60 ⁇ m.
  • Capture rate is defined as (captured biological substance/original biological substance in the testing sample) ⁇ 100%.
  • Channel O has no microstructure and has the lowest biological substance capture rate, at 27% and 1% for DMEM sample and blood sample, respectively.
  • Design E has a 80% capture rate for HCT116 cancer cells spiked in DMEM, and a 30% capture rate for HCT116 cancer cells spiked in blood sample.
  • Design F has the best capture rate, on average over 70% of HCT116 cancer cells spiked in blood sample were captured (see FIG. 7J ).
  • the biological substance on the surface coating can be further purified by removing the non-specific cells and other blood components on the surface of the nonfouling/releasable composition.
  • the nonfouling/releasable composition has low affinity for non-specific cells and other blood components. Therefore, rinsing the surface coating with a low flow buffer solution of about 0.8 dyne/cm 2 to about 50 dyne/cm 2 is sufficient to remove non-specific cells and other blood components on the nonfouling/releasable composition while the biological substance remains on the surface coating.
  • the shear force of the buffer rinse is about 2.5 to about 10 dyne/cm 2 .
  • FIG. 8 shows that when the shear stress of the buffer flow is about 3.3 dyne/cm 2 , 80% of the non-specific cells (i.e. white blood cells) were removed while none of the biological substance (i.e. HCT 116 cancer cells) were removed from the surface coating.
  • the shear stress of the buffer flow was increased to 8 dyne/cm 2 , almost all of the non-specific cells were removed while none of the biological substance was removed from the surface coating.
  • the, biological substance After removing the majority of the non-specific cells and blood components by flow purification, the, biological substance can be released from the surface coating.
  • the captured biological substance can be released by introducing an air bubble solution or oil phase.
  • the surface coating comprises a nonfouling composition A (lipid bilayer) and a bioactive composition B (antibody) and is bound to a solid substrate S.
  • the biological substance, CTC is bound to the bioactive composition B, whereas other cells were repelled by the nonfouling composition A.
  • the hydrophobic tails of the lipid bilayer are turned upside down due to its high affinity with the air inside the air bubble, which is also hydrophobic. This breaks up the hydrophilic-hydrophilic interaction at the surface of the lipid Hayes and allows the air bubble to “lift off” the top layer of the lipid bilayer, together with the CTC bound on the bioactive composition.
  • the captured biological substance can be released by breaking the cleavable functional group on the linker composition or on the surface linker.
  • This release mechanism is illustrated in FIGS. 10A and 10B .
  • FIG. 10A shows a surface coating on a solid substrate, wherein the surface coating comprises a bioactive composition B, a linker composition with a cleavable functional group C, and a nonfouling composition A.
  • the surface coating is attached to a solid substrate S (e.g. glass) by a surface linker 1 .
  • FIG. 10B shows the release of the biologic substance (e.g. CTC) from the surface coating in FIG. 10A .
  • the biologic substance e.g. CTC
  • the biologic substance is bound to the bioactive composition B, whereas other cells were repelled by the nonfouling composition A.
  • the surface coating is irradiated with 365 nm ultraviolet light, which breaks the cleavable functional group on the linker composition C and the biologic substance is released for subsequent analysis but maintaining the viability.
  • the biological substance can also be released by other mechanisms.
  • the linker composition or the surface linker comprises an electrosensitive cleavable functional group, and the biological substance is released by electro pulse mechanism.
  • the linker composition or the surface linker comprises a magnetic material as the cleavable functional group, and the absence of the magnetic field or force releases the biological substance.
  • the linker composition or the surface linker comprises a PEM as the cleavable functional group, and the biological substance is released by changing the electrostatic interaction between the layers.
  • the linker composition or the surface linker comprises an DNA piece as the cleavable functional group, and the biological substance is released by DNA hybridization.
  • Supported lipid bilayer was prepared by the following steps:
  • Biotinylated EpCAM Antibody was prepared by the following steps:
  • biotinylated goat anti-human anti-EpCAM antibody from R and D Systems (Minneapolis, Minn.) could be used.
  • Glass substrate such as microscope coverslips from Deckglaser, Germany
  • 10% DECON 90 Devon Laboratories Limited, England
  • Milli-Q water dried under nitrogen gas
  • oxygen plasma in a plasma cleaner (Harrick Plasma, Ithaca, N.Y., U.S.A.) at 100 mtorr for 10 min.
  • a plasma cleaner Hard Plasma, Ithaca, N.Y., U.S.A.
  • Silicon oxide based solid substrates e.g. silicon wafer or glass coverslips
  • piranha solution 70% sulfuric acid and 30% hydrogen peroxide (v/v)
  • piranha solution 70% sulfuric acid and 30% hydrogen peroxide (v/v)
  • v/v hydrogen peroxide
  • SA streptavidin
  • Si-bPEGs The biotinylated PEG si lane solution (Si-bPEGs) was added to the clean glass substrate and incubated for 1 hour to form a Si-bPEG nonfouling composition on the glass substrate, followed by an ethanol rinse to remove excess Si-bPEGs.
  • Silane was the surface linker and the biotin was the functional group that bind with the functional group (SA) in the linker composition.
  • b-Anti-EpCAM solution 0.05 mg/mL was added and bound with SA-Si-bPEGs surface coating, followed by PBS buffer rinse to remove excess b-Anti-EpCAM.
  • PLL MW 15000-30000; Sigma, St Louis, Mo.
  • Tris-HCl buffer 1 mg/mL
  • PLL-coated slide was subsequently immersed in the PLGA solution (MW 3000-15000, Sigma, St Louis, Mo., 1 mg/mL) for 10 min, followed by rinsing with 1 mL of Tris-HCl buffer for 1 min.
  • c-(PLL/PLGA)i where i was denoted as the number of polyelectrolyte pairs generated by repeating the above steps: i) 0.5 was referred to c-PLL only, i) I was referred to c-(PLL/PLGA)1, and the like.
  • the construction of the surface coating was monitored by quartz crystal microbalance with dissipation (QCM-D).
  • QCM-D quartz crystal microbalance with dissipation
  • FIG. 11 shows the construction of the surface coating on a SiO 2 -pretreated quartz crystal.
  • the characteristics of the SLB nonfouling composition on the surface coating were examined using QCM-D ( FIG. 12 ).
  • microfluidic p can be prepared by the following steps:
  • Each blood sample contained 2 ml of blood from a stage IV colon cancer patient and the sample was introduced to the sealed channel of the microfluidic chip at 0.5 ml/hr, controlled by a syringe pump. Subsequently, the sealed channel in the microfluidic chip was rinsed with 0.5 ml of PBS buffer at the flow rate of 1 ml/h, followed by in situ immunostaining.
  • the number of CTCs captured per ml of blood for these 8 samples were 26, 34, 36, 39, 47, 67 79, and 99. 25% of the blood samples had 79 or higher CTC count per ml of testing sample and the median CTC count was 43 per ml of testing sample. There was minimal binding of the non-specific cells and proteins after the buffer rinse.
  • the CTC count for the FDA approved Veridex CellSearch is as follows: 25% of the samples had 3 or more CTCs per 7.5 ml of testing sample and the median CTC counts was 0.
  • the anti-EpCAM functionalized SLB surface was incubated with 150 uL of HCT116 cancer cell spiked human blood (with HCT116 cancer cell density of approximately 10 to 100 per 100 ⁇ L of blood), followed by a buffer rinse to remove non-specific cells.
  • FIG. 13 shows the surface coating before and after the buffer rinse. Prior to the buffer rinse, the surface coating was covered with non-specific cells (upper left) and four HCT116 cancer cells (lower left). After the buffer rinse, almost all of the non-specific cells were removed (upper right) but the four HCT116 cancer cell (lower right) remained on the surface coating.
  • the results show the surface coating of the present invention effective in capturing CTCs and releasing the non-specific cells.
  • HCT116 cancer cells biological substance
  • nonfouling property of six different surface conditions are illustrated in FIG. 14A .
  • FIG. 14B shows the non-specific, blood cell binding of the following surfaces: (A) Glass only; (B) biotinylated SLB (b-SLB), (C) Streptavidin conjugated-bSLB, and (D) OC98-1-conjugated bSLB. These surfaces were incubated with diluted human blood from healthy donor (1 uL, of blood in 100 uL PBS buffer) for 4 hours, followed by a PBS buffer rinse. Images (E) to (H) are the after rinse images which correspond to the surface coatings in (A) to (D). The results show that after a buffer rinse, there is less non-specific blood cell on the surface coatings with a releasable composition (i.e. SLB) compare to the surface coating without a releasable composition (i.e. glass only).
  • a releasable composition i.e. SLB
  • the differentiated flow shear could selectively “flush” out the non-specific cells based on the affinity of these cells to the nonfouling composition, while the biological substance remains on the surface coating.
  • FIG. 15A shows fibroblast 3T3 (green) and colon cancer cell line HCT116 (red) were incubated on the surface coating for 4 h.
  • the surface coating was rinsed with a buffer solution, which has a shear stress of 3 dyne/cm 2 .
  • the HCT 116 cells (red) were flushed away from the surface coating within 5 min of the buffer rinse, as shown in FIG. 15B .
  • the fibroblast 3T3 cells (green) remained on the surface coating after 30min of buffer rinse, as shown in FIG. 15C , due to its high affinity to fibronectin.
  • the result shows a shear stress about 3 dyne/cm 2 is sufficient to remove the non-specific cells from the releasable composition.
  • FIG. 16 summarizes the respective shear stress and flushing time for the HCT116 and NIH-3T3 cell populations (non-specific cells).
  • the shear stress is about 3 to about 4.5 dyne/cm 2 .
  • the shear stress is about 8.5 to about 12 dyne/cm 2 (N/N0 is the percentage of the cells remains attached to the surface coating using various shear stresses, N is the final cell number and N0 is the initial cell number.)
  • FIG. 17 shows HCT116 cells in the red circle were removed from the surface coating within 3 seconds of introducing air bubbles.
  • the captured CTCs were incubated with 5 mM of EDTA at 37° C. for 5 to 10 min and released by flowing a culture medium into the sealed channel of the microfluidic chip. A total of 18 colo205 cells were released from this procedure. The released colo205 cells, together with a serum-containing culture medium and antibiotics (penicillin+streptomycin+gentamicin), were placed into a 48-well tissue cultured polystyrene plate for cultivation.
  • antibiotics penicillin+streptomycin+gentamicin
  • FIGS. 18A-18C show a portion of 18 colo205 cells on day 1, on day 10 and day 14 respectively. This study demonstrates the released colo205 cells retained their viability for subsequent cell culture.
  • FIG. 19 illustrates schematically a filtration device, wherein the filtration filter is coated with the surface coating of the present invention.
  • the filter could accommodate high volume blood flow and capture a biological substance for a diagnostic or therapeutic purpose.
  • a catheter can be inserted into the patient's vein or fistula and the patient's blood flows through the CTC filtration device, wherein the surface coating on the filters captures the CTCs. The filtered blood flows back to the patient.
  • biotinylated OC9801 antibody biotinylated EpAb4-1 antibody
  • biotinylated EpCam antibody commercially available from R&D system, USA
  • the CTCs were spiked in a buffer solution (about 10 5 CTCs/ ml).
  • the CTC-spiked buffer solution was introduced to the surface coatings with the following bioactive composition: biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody, biotinylated EpCam antibody and IgC1 antibody.
  • FIG. 20 shows biotinylated EpAb 4-1 is effective in capturing HCT116 CTCs and SAS CTCs.

Abstract

This invention relates to a surface coating for capture circulating rare cells, comprising a nonfouling composition to prevent the binding of non-specific cells and adsorption of serum components; a bioactive composition for binding the biological substance, such as circulating tumor cells; with or without a linker composition that binds the nonfouling and bioactive compositions. The invention also provide a surface coating for capture and purification of a biological substance, comprising a releasable composition to release the non-specific cells and other serum components; a bioactive composition for binding the biological substance, such as circulating tumor cells; with or without a linker composition that binds the releasable and bioactive compositions. The present invention also discloses a novel microfluidic chip, with specific patterned microstructures to create a flow disturbance and increase the capture rate of the biological substance.

Description

    CROSS-REFERENCES TO RELATED APPLICATIONS
  • This application is a nonprovisional application of a U.S. Patent Application Ser. No. 61/502,844, filed on 29 Jun. 2011, and U.S. Patent Application Ser. No. 61/606,220, filed on 2 Mar. 2012, which are incorporated by reference in their entirety.
  • STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
  • NOT APPLICABLE
  • REFERENCE TO A “SEQUENCE LISTING,” A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK
  • Table 1 is the amino acid sequence of EpAb4-1 antibody.
  • BACKGROUND OF THE INVENTION
  • The shedding of cells into the circulation is an intrinsic property of the malignant tumor, and this feature provides important information with regard to the diagnosis, staging, treatment response and survival of cancer patients. For example, Pantel et al found the number of circulating tumor cells (CTCs) in the blood is correlated with the aggressiveness of the cancer as well as the efficacy of the therapy. (Pantel, K. et. al., “Detection, clinical relevance and specific biological properties of disseminating tumor cells”, Nat Rev Cancer, 2008, 8(5):329-40).
  • However, CFCs, as few as one per 109 blood cells in patients with metastatic cancer, are rare cells. This makes the detection and isolation of CTCs technically challenging (sec Kahnet al. Breast Cancer Res Treat 2004, 86:237-47). An enrichment process is therefore necessary to effectively detect and isolate CTCs.
  • An example of such enrichment process is the use of a highly overexpressed cell surface biomarker with high specificity and sensitivity for CTCs, such as the epithelial cell adhesion molecule (EpCAM). The Cellsearch System™ (Veridex), the only FDA-approved platform for CTC detection, utilizes anti-EpCAM antibody-coated magnetic nanoparticles to capture and enrich CTCs, followed by cytokeratin immunostaining. The AdnaTest (AdnaGen AG, Germany), another commercially available system for CTC detection, adopts similar immunomagnetic approach by using anti-EpCAM and Mucin 1 (MUC1) conjugated magnetic beads. More recently, “CTC chips” based on anti-EpCAM antibody-coated microfluidics chip were developed for CTC detection and enrichment (Nagrath et al, Nature 2007, 450:1235-9). However, the disadvantage of the above techniques is the low detection rate of pure CTCs, due to the non-specific binding of blood cells with anti-EpCAM antibody.
  • In order to maximize the detection and isolation of CTCs, it is necessary to reduce the nonspecific binding of other circulating blood cells. This can be achieved by surface modification with bioinert materials. For example, Kaladhar et al. observed a significant fewer circulating blood cells (e.g. platelets, leukocytes, and erythrocytes) binding onto the solid substrate modified with supported monolayer of various lipid compositions containing phosphatidyl-choline, cholesterol, and glycolipid (Kaladhar et al, Langmuir 2004, 20: 11115-22 and Kaladhar et al, J Biomed Mater Res A 2006, 79A:23-35).
  • Despite the advance in the detection and isolation CTCs technology, there is still a need for a more specific and effective method for detecting, purification and releasing; CTCs and other biological substances for further cultivation and characterization.
  • BRIEF SUMMARY OF THE INVENTION
  • In one aspect, the present invention is directed to a surface coating to capture a circulating rare cell (CRC). This surface coating increases the capture efficiency of a CRC, such as CTC, circulating stem cells (e.g. tumor stem cell and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like and reduces the binding of non-specific cells or protein adsorption.
  • The surface coating comprises 1) a nonfouling composition that reduces the binding of nonspecific blood cells and adsorption of other blood components, such as protein; and 2) a bioactive composition that captures a CRC. The surface coating further comprises a linker composition that attaches to the nonfouling composition and the bioactive composition, as illustrated in FIG. 1A.
  • In another aspect, the present invention is directed to a surface coating to capture and release a biological substance. This surface coating increases the capture efficiency of a biological substance, such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like and enhances the removal or release of the non-specific cells or protein from the surface coating.
  • The surface coating comprises 1) a releasable composition for releasing or removing nonspecific blood cells and other blood components, such as protein, from the surface coating; and 2) a bioactive composition that captures a biological substance. The surface coating further comprises a linker composition that attaches to the releasable composition and the bioactive composition.
  • The present invention is also directed to a microfluidic device, with specific microstructure designs to create a disturbed flow of blood, body fluid or biological samples to increase the capture rate of the biological substance.
  • The present invention is also directed to a method of manufacturing the surface coating, comprising a) forming the nonfouling or the releasable composition; and b) attaching the the linker composition with the nonfouling/releasable composition from step a) and the bioactive composition, or c) attaching the nonfouling/releasable composition from step a) with the bioactive composition.
  • The present invention is also directed to methods to capture and release the biological substance from the surface coating. The biological substance on the surface coating can be purified by removing the non-specific cells or protein. The captured biological substance can be released by air bubbles, ultraviolet irradiation and the like.
  • The present invention is also directed to uses of a biotinylated anti-EpCam antibody, EpAb4-1 antibody, to capture a CTC.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • Embodiments of the present invention may be described with reference to the accompanying drawings.
  • FIG. 1A illustrates schematically an embodiment of the surface coating comprising a nonfouling composition, a linker composition and a bioactive composition.
  • FIG. 1B illustrates schematically the binding of a circulating tumor cell with the surface coating from FIG. 1A
  • FIG. 2A to FIG. 2F illustrate the chemical structures of examples of nonfouling materials.
  • FIG. 3 illustrates the chemical reactions of conjugation between the functional groups on the nonfouling composition and the bioactive composition.
  • FIG. 4A illustrates schematically the attachment of the surface coating and solid substrate without a surface linker.
  • FIG. 4B and FIG. 4C illustrate schematically a linker composition with a cleavable functional group.
  • FIG. 4D illustrates schematically the attachment of the surface coating and the solid substrate using a surface linker.
  • FIG. 5A and FIG. 5B illustrates schematically the formation of the surface coating on a solid substrate.
  • FIGS. 6A and 6B illustrate schematically the components of a microfluidic chip.
  • FIG. 6C illustrates schematically the microfluidic chip assembly to capture CTCs from a biological sample.
  • FIG. 7A to FIG. 7H illustrate schematically the designs of the microstructures the solid substrate.
  • FIGS. 7I and 7J illustrate the capture efficiency of various microstructure designs in DMEM solution and blood respectively.
  • FIG. 8 illustrates the shear stresses of a buffer solution to release the non-specific cells and purify the captured biological substance.
  • FIG. 9. illustrates schematically the release of biological substance by the air bubble method.
  • FIG. 10A illustrates schematically lie surface coating with a cleavable linker composition on a solid substrate.
  • FIG. 10B illustrates schematics the release of the biologic substance from the surface coating in FIG. 10A.
  • FIG. 11 illustrates QCM-D response of the surface coating construction.
  • FIG. 12 illustrates the QCM-D response of the addition of bovine serum albumin the surface coating.
  • FIG. 13 are the photographs of the non-specific cells (top images) and the CTCs (bottom images) on the surface coating before and after the buffer rinse.
  • FIG. 14A illustrates the capture efficiency and non-specific blood cell binding of various surface coatings.
  • FIG. 14B are photo images which illustrate the non-specific blood cell binding of various surface coatings before and after the buffer rinse.
  • FIG. 15A to FIG. 15C are the photographs of the non-specific cells and the biological substance on the surface coating before and after the buffer rinse purification.
  • FIG. 16 illustrates the various shear stress and flushing time for the removal of HCT116 and NTH-3T3 cell populations from the surface coating.
  • FIG. 17 are the photographs of the CTCs released by the air bubbles.
  • FIG. 18 illustrates the cell cultures of the released CTCs on day 1day 10 and day 14.
  • FIG. 19 illustrates schematically a CTC filtration device.
  • FIG. 20 illustrates the CTC binding specificity of biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody, biotinylated EpCam antibody and IgG antibody,
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to a surface coating to effectively capture a circulating rare cell (CRC), such as CTC, circulating stem cells (e.g. tumor stem cell and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like.
  • In one embodiment, the surface coating for the capture of a CRC comprises 1) a nonfouling composition that prevents the binding of non-specific cells and adsorption of other blood components, such as protein; and 2) a bioactive composition that captures the circulating rare cells. The nonfouling composition and the bioactive composition are joined by discrete functional groups or moieties present in the nonfouling and bioactive compositions. Generally, a linkage between the two compositions is formed by an interaction comprising electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, complementary DNA binding, magnetic force, or combinations thereof.
  • In one group of embodiments, complementary DNA fragments are used for binding the nonfouling composition and the bioactive composition. The fragments are attached to each of the compositions and can be partially or completely complementary over their lengths. A suitable length of DNA will generally be at least 15, 20, 25, 35, 50, 100 or more bases in length. An example of the DNA used in the present invention is an DNA tweezer. (See, B Yurke et al., A DNA-fuelled molecular machine made of DNA. Nature 2000, 406:605-608.)
  • In another group of embodiments, the surface coating comprises 1) a nonfouling composition; 2) a bioactive composition; and 3) a linker composition, which joins the nonfouling composition to the bioactive composition. See FIG. 1A.
  • The present invention is also directed to a surface coating to effectively capture a biological substance, such as CTC, circulating stem cells (e.g. tumor stem cell, liver stem cells and bone marrow stem cells), fetal cells, bacteria, virus, epithelial cells, endothelial cells or the like, purify the biological substance on the surface of the surface coating by releasing or removing the non-specific cells and other serum components (e.g. protein) through a buffer rinse, and release the captured biological substance from the surface coating.
  • The surface coating for the capture and purification of a biological substance comprises 1) a releasable composition for releasing nonspecific blood cells and other blood components, such as protein, through a buffer rinse; and 2) a bioactive composition that captures a biological substance. The releasable composition and the bioactive composition are joined by discrete functional groups or moieties present in the releasable and bioactive compositions. Generally, a linkage between the two compositions is formed by an interaction comprising electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, complementary DNA binding, magnetic force, or combinations thereof.
  • In one embodiment, the surface coating further comprises a linker composition that attaches to the releasable composition and the bioactive composition.
  • As will be explained in more detail below, the surface coating can be incorporated into the following configurations: cell cultural dishes, microfluidic channels, microfluidic chips, filtration filter, capillaries, tubes, beads, nanoparticies, or the like, with an inner diameter ranging from about 50 to about 1000 um.
  • Nonfouling and Releasable Composition
  • “nonfouling” composition (see FIG. 1A) reduces the binding of non-specific cells and adsorption of the serum protein.
  • The “releasable” composition comprises a nonfouling composition which also acts as a “lubricating” surface such that only low flow shear stress is required to remove or release the non-specific cells or blood components from the surface coating, while the biological substance remains intact.
  • The nonfouling composition is selected from the group consisting of: a supported lipid layer such as liposomes, supported lipid bilayers (SLBs) or lipid multilayer, polypeptides, polyelectrolyte multilayers (PEMs), polyvinyl alcohol, polyethylene glycol (PEG) as illustrated in FIG. 2A, hydrogel polymers, extracellular matrix proteins, carbohydrate, polymer brushes, zwitterionic materials such as poly(carboxybetaine) (pCB)) as illustrated in FIG. 2D, poly(sulfobetaine) (pSB) as illustrated in FIG. 2E and pDMAEMA as illustrated in FIG. 2F, small organic compounds, and the combination of above materials forming a single or a multi-layer.
  • For those embodiments in which the nonfouling composition comprises supported lipid bilayers (SLBs), the SLBs typically comprise lipids such as, for example, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(cap biotinyl) (sodium salt) (b-PE) as illustrated in FIG. 2B and 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC). The protein resistant property of a SLB can be explained by the presence of neutral and zwitterionic phosphatidylcholine headgroups in a wide pH range, as well as an aqueous thin film formed between the hydrophilic lipid head groups and the bulk solution (see, Johnson et al., Biophys J 1991, 59:289-94).
  • In another group of embodiments, the nonfouling composition comprises PEG, preferably PEG with a molecular weight from about 100 to about 100,000 and exhibits a nonfouling property.
  • In yet another group of embodiments, the nonfouling composition comprises polyelectrolyte multilayers (PEMS) or a polymer brush. Examples of suitable PEMs useful in the present invention include, but are not limited to, poly-L-lysine/poly-L-glutamine acid (PLL/PLGA), poly-L-lysine/poly-L-aspartic acid or similar counter ionic polyelectrolytes. The polymer brush comprises ([2-(acryloyloxy)ethyl] trimethyl ammonium chloride, TMA)/(2-carboxy ethyl acrylate, CAA) copolymer as illustrated in FIG. 2C. Generally, the nonfouling layer has a thickness from a few nanometers up to hundreds microns.
  • The nonfouling composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment, either directly to a functional group present in the bioactive composition, or directly to a functional group that is part of the linkage composition.
  • In some embodiments, the functional groups of the nonfouling composition (prior to covalent attachment) are selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the linker or bioactive composition. In other embodiments, the functional groups of the nonfouling composition (prior to non-covalent attachment) which are first members of a binding pair, are selected from the group using specific binding recognition consisting of avidin, streptavidin DNA, RNA, ligand, receptor, antigen, antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present in either the linker or bioactive composition.
  • The Linker Composition
  • The linker composition joins the nonfouling/releasable composition and the bioactive composition and comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling/releasable composition and to a functional group that is part of the bioactive composition.
  • In some embodiments, the linker composition comprises functional groups (prior to covalent attachment) selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups, which are selected to be reactive with functional groups present in either the nonfouling or bioactive composition.
  • In other embodiments, the linker composition comprises functional groups (prior to non-covalent attachment) which are first members of a binding pair, selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen, antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present on the nonfouling/releasable composition or the bioactive composition.
  • The functional groups on the linker composition can also be a cleavable functional group, selected from: a photosensitive functional group cleavable by ultraviolet irradiation, an electrosensitive functional group cleavable by electro pulse mechanism, a magnetic material cleavable by the absence of the magnetic force, a polyelectrolyte material cleavable by breaking the electrostatic interaction, a DNA cleavable by hybridization, and the like.
  • Bioactive Composition
  • The bioactive composition joins to either the linker composition or the nonfouling composition, and comprises a binding moiety selective for the detection of the biological substance or CRC.
  • The bioactive composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling layer or to a functional group that is part of the linker composition.
  • In some embodiments, the functional groups of the bioactive composition (prior to covalent attachment) are selected from: hydroxy groups, amine groups, carboxylic acid or ester groups, thioester groups, aldehyde groups, epoxy or oxirane groups, hyrdrazine groups and thiol groups which are selected to be reactive with functional groups present in either the nonfouling or linker composition. In other embodiments, the functional groups of the bioactive composition (prior to non-covalent attachment) are selected from the group using specific binding recognition consisting of biotin, avidin, streptavidin, DNA, RNA, ligand, receptor, antigen--antibody and positive-negative charges, each of which is selected to bind to a second member of the binding pair which is present on the nonfouling/releasable composition or the linker composition.
  • The binding moiety of the bioactive composition has specific affinity with the biological substance through molecular recognition, chemical affinity, or geometrical/shape recognition. Examples of the binding moiety for the detection of the biological substance include, but are not limited to: synthetic polymers, molecular imprinted polymers, extracellular matrix proteins, binding receptors, antibodies, DNA, RNA, antigens or any other surface markers which present high affinity to the biological substance. A preferred antibody is the anti-EpCAM membrane protein antibody (commercially available from many sources, including R&D Systems, MN, USA), which provides high specificity for CTCs because EpCAM is frequently overexpressed in the lung, colorectal, breast, prostate, head and neck, and hepatic malignancies, but is absent from haematologic cells. Another preferred antibody is Anti-HER2, which has high specificity for CFCs but absent in haematologic cells.
  • In one embodiment, the anti-EpCAM membrane protein antibody is EpAb4-1 antibody, comprising a heavy chain sequence with SEQ ID No:1 and a light chain sequence with SEQ ID NO: 2 shown in Table 1.
  • TABLE 1
    Amino Acid Sequence of VH and VL domains of EpAb4-I antibody
    FW1 CDR1 FW2 CDR2
    SEQ ID QIQLVQSGPELKKPGETV GYTFTNYG WVKQAPGKGLK INTYTGEP
    NO: 1 KISCKAS MN WMGW
    (VH)
    SEQ ID DIVMTQAAFSNPVTLGTS RSSKSLLH WYLOKPGQSPQ HMSNLAS
    NO: 2 ASISC SNGITYLY LLIY
    (VL)
    FW3 CDR3 FW4 Family
    SEQ ID TYGDDFKGRFAFSLETSA FGRSVDF WGQGTSVTVSS VH9
    NO: 1 STAYLQINNLKNEDTATY
    (VH) FCAR
    SEQ ID GVPDRFSSSGSGTDFILRI AQNLENPR FGGGTKLEIK VK24/25
    NO: 2 SRVEAEDVGIYYC T
    (VL)

    Complementary-determining regions 1-3 (CDR1-3), framework regions 1-4 (FW1-4) for both the VH and VL domains are shown. The V domain families were aligned by VBASE2 database www.vbase2.org).
  • The bioactive composition can have a variety of thicknesses, selected so that it does not affect the function or the performance of the surface coating.
  • In one embodiment, the conjugation linkers or catalysts for the nonfouling composition and the bioactive compositions are biotin/avidin or their derivatives. In another embodiment, the conjugation linkers or catalysts for the nonfouling composition and the bioactive composition are EDC/NHS. In yet another preferred embodiment, the conjugation linkers or catalysts for the nonfouling composition and the bioactive compositions are sulfo-SMCC. FIG. 3 schematically illustrates the chemical reactions of these embodiments.
  • Solid Substrate
  • In some embodiments, the surface coating is attached to the solid substrate without a surface linker, as illustrated in FIG. 4A. The nonfouling/releasable composition is attached to the solid substrate via one of the following interactions: covalent bonding (for PEG nonfouling composition), hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction (for SLB nonfouling/releasable composition), polar-polar interaction, complimentary DNA binding, magnetic force, or the like.
  • In other embodiments, the surface coating is attached to the solid substrate with a surface linker, as illustrated in FIG. 4D. Examples of the solid substrate used in the present invention include, but are not limited to: metals, plastics, glass, silicon wafers, hydroxylated poly(methyl methacrylate) (PMMA), and a combination thereof. The shape of the solid substrate include, but are not limited to: planar, circular and irregular shapes with micro, or nano-structures such as nanoparticles, nanowires, and a combination thereof.
  • The surface linker composition comprises functional groups capable of covalent, non-covalent, or a combination of covalent and non-covalent attachment directly to a functional group present in the nonfouling/releasable composition and to a functional group that is part of the solid substrate. Examples of the surface linker for binding the surface coating to a glass substrate include, but are not limited to, silane, aminopropyltriethoxy aminopropyltrimethoxy silane, silane-PEG-NH2, silane-PEG-N3 (PEG molecular weight is about 1,000 to about 30,000 daltons) and silane-PEG biotin.
  • In one group of embodiments, the surface linker comprises a cleavable functional group selected from: a photosensitive functional group cleavable by ultraviolet irradiation, an electrosensitive functional group cleavable by electro-pulse mechanism, an iron or magnetic material in which the absence of the magnetic force will release the nonfouling composition a polyelectrolyte material cleavable by breaking the electrostatic interaction, an DNA cleavable by hybridization, and the like.
  • In one embodiment, the nonfouling composition comprises silane-functionalized PEG and the solid substrate is preferably selected from the group consisting of silicon, glass, hydroxylated poly(methyl methacrylate) (PMMA) aluminum oxide, TiO2 and the like. In another embodiment, the nonfouling composition comprises thiol-functionalized compounds and the solid substrate is preferably selected from the group consisting of Au, Ag, Pt, and the like.
  • The Method of Manufacturing the Surface Coating
  • FIGS. 5A and 5B show the steps of forming the surface coating:
    • 1. Formation of the nonfouling/releasable composition (e.g. SLB or PEG) with appropriate functional group (biotin);
    • 2. Attaching the functional group (streptavidin) on the linker composition to the functional group (biotin) on the nonfouling/releasable composition;
    • 3. Formation of the bioactive composition and attaching the functional group (biotin) on the bioactive composition to the functional group (streptavidin) on the linker composition.
  • he surface coating without a linker composition can be formed by:
    • 1. Formation of the nonfouling/releasable composition with appropriate functional group (e carboxyl group of N-glutaryl phosphatidylethanolamine or NGPE);
    • 2. Formation and attaching the functional group (primary amine) on the bioactive composition to the functional group (carboxyl group of NGPE) on the nonfouling/releasable composition in step 1.
  • The steps in paragraphs [0077] and [0078] can be reversed.
  • Microfluidic Chip
  • As illustrated in FIG. 6A, the microfluidic chip comprises a first solid substrate 1 (e.g. PMMA) and a second solid substrate 2 (e.g. glass), wherein the first and second solid substrates are adhered together using an adhesive means 3 or other means.
  • Referring to FIG. 6B, the surface of one or both solid substrates can be engraved with microstructures 4. In one group of embodiments, the microstructures 4 are arranged in a linear fashion. In another group of embodiments, the microstructures 4 are arranged in herringbone fashion. The shaded region on the adhesive 3 in FIG. 6B is carved out to accommodate the microstructures 4 on the surface of the solid substrate 1. A sealed channel 5 is created by adhering the first solid substrate 1 and the second solid substrate 2 together with an adhesive 3. The height of the channel 5 is determined by the thickness of the adhesive 3.
  • Once the microfluidic chip is formed, the surface coating can be attached to one or both solid substrates. In one group of embodiments, the surface coating is attached to the solid substrate with a surface linker. In another group of embodiments, the surface coating is attached to the solid substrate via one of the following interactions: covalent bonding (for PEG nonfouling composition), hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction (for SLB nonfouling/releasable composition), polar-polar interaction, complimentary DNA binding, magnetic force, or the like.
  • Referring to FIG. 6C, the microstructures 4 on the solid substrate, arc perpendicular to the flow direction and create a chaotic or disturbed flow of the blood, body fluid or biologic sample as it passes through the sealed channel 5 of the microfluidic chip. The disturbed flow enhances the biological substance-surface coating contact.
  • Two factors govern the capture efficiency of the microfluidic chip:
    • (1) The linear speed of the blood, body fluid or biological sample, which determines the contact time of the biological substance and the surface coating. In a preferred embodiment, the linear speed is about 0.1 mm/s to 1 mm/s. In a more preferred embodiment, the linear speed is about 0.42 mm/s or 0.5 ml/h for Design E in FIG. 7F.
    • (2) The flow disturbance of the blood, body fluid or biological sample, created by the microstructures 4 on the solid substrate(s). The flow disturbance increases contact between the biological substance and the surface coating.
  • FIG. 7A shows various designs of the microstructures 4 on the solid substrate. The microstructures in Design F are arranged in a herringbone pattern whereas the microstructures in Designs A-E and H are arranged in a linear pattern. The dimensions of the microstructures 4 are as follows: the length is about 50 mm fir O-D and G and about 120 mm for E-F, the height is about 30 μm, the width is about 1.5 mm for O and A, about 3.0 mm for B, and about 5..5 mm for C-G. The height of the sealed channel 5 varies with the thickness of the adhesive 3, preferably about 30-90 μm, more preferably about 60 μm.
  • FIG. 7B-7H show the details of Designs A-C in FIG. 7A. Design C in FIG. 7H is the preferred pattern, with the following dimensions: the width of Microstructure (W) is about 150 μm, the length of microstructure (L) is about 1000 μm, the distance between two rows of microstructures (Sr) is about 250 μm, the distance between two adjacent microstructures (Sc) is about 350 μm, the height of the microstructure (D) is about 30 μm and the height of the sealed channel 5 (H) is about 60 μm.
  • The biological substance capture efficiency of the various designs are shown in FIG. 7I and FIG. 7J. Capture rate is defined as (captured biological substance/original biological substance in the testing sample)×100%. Channel O has no microstructure and has the lowest biological substance capture rate, at 27% and 1% for DMEM sample and blood sample, respectively. Design E has a 80% capture rate for HCT116 cancer cells spiked in DMEM, and a 30% capture rate for HCT116 cancer cells spiked in blood sample. Design F has the best capture rate, on average over 70% of HCT116 cancer cells spiked in blood sample were captured (see FIG. 7J).
  • Flow Purification
  • The biological substance on the surface coating can be further purified by removing the non-specific cells and other blood components on the surface of the nonfouling/releasable composition. The nonfouling/releasable composition has low affinity for non-specific cells and other blood components. Therefore, rinsing the surface coating with a low flow buffer solution of about 0.8 dyne/cm2 to about 50 dyne/cm2 is sufficient to remove non-specific cells and other blood components on the nonfouling/releasable composition while the biological substance remains on the surface coating.
  • In a preferred embodiment, the shear force of the buffer rinse is about 2.5 to about 10 dyne/cm2. FIG. 8 shows that when the shear stress of the buffer flow is about 3.3 dyne/cm2, 80% of the non-specific cells (i.e. white blood cells) were removed while none of the biological substance (i.e. HCT 116 cancer cells) were removed from the surface coating. When the shear stress of the buffer flow was increased to 8 dyne/cm2, almost all of the non-specific cells were removed while none of the biological substance was removed from the surface coating.
  • Release of the Biological Substance
  • After removing the majority of the non-specific cells and blood components by flow purification, the, biological substance can be released from the surface coating.
  • If the nonfouling/releasable composition comprises a lipid or a mixture of lipid, the captured biological substance can be released by introducing an air bubble solution or oil phase. As shown in FIG. 9, the surface coating comprises a nonfouling composition A (lipid bilayer) and a bioactive composition B (antibody) and is bound to a solid substrate S. The biological substance, CTC, is bound to the bioactive composition B, whereas other cells were repelled by the nonfouling composition A. As the air bubble approaches the lipid bilayer, the hydrophobic tails of the lipid bilayer are turned upside down due to its high affinity with the air inside the air bubble, which is also hydrophobic. This breaks up the hydrophilic-hydrophilic interaction at the surface of the lipid Hayes and allows the air bubble to “lift off” the top layer of the lipid bilayer, together with the CTC bound on the bioactive composition.
  • If the nonfouling compost ion comprises a composition other than a lipid or a mixture of lipid, the captured biological substance can be released by breaking the cleavable functional group on the linker composition or on the surface linker. This release mechanism is illustrated in FIGS. 10A and 10B. FIG. 10A shows a surface coating on a solid substrate, wherein the surface coating comprises a bioactive composition B, a linker composition with a cleavable functional group C, and a nonfouling composition A. The surface coating is attached to a solid substrate S (e.g. glass) by a surface linker 1. FIG. 10B shows the release of the biologic substance (e.g. CTC) from the surface coating in FIG. 10A. The biologic substance is bound to the bioactive composition B, whereas other cells were repelled by the nonfouling composition A. The surface coating is irradiated with 365 nm ultraviolet light, which breaks the cleavable functional group on the linker composition C and the biologic substance is released for subsequent analysis but maintaining the viability.
  • The biological substance can also be released by other mechanisms. In one group of embodiments, the linker composition or the surface linker comprises an electrosensitive cleavable functional group, and the biological substance is released by electro pulse mechanism. In another group of embodiments, the linker composition or the surface linker comprises a magnetic material as the cleavable functional group, and the absence of the magnetic field or force releases the biological substance. In yet another group of embodiments, the linker composition or the surface linker comprises a PEM as the cleavable functional group, and the biological substance is released by changing the electrostatic interaction between the layers. In yet another group of embodiments, the linker composition or the surface linker comprises an DNA piece as the cleavable functional group, and the biological substance is released by DNA hybridization.
  • EXAMPLES
  • The following examples further illustrate the present invention. These examples are intended merely to be illustrative of the present invention and are not to be construed as being limiting.
  • Example 1 Preparation of the Two-Layer Surface Coating Preparation of the Nonfouling Composition:
  • Supported lipid bilayer (SLB) was prepared by the following steps:
    • (1) POPC and b-PE (commercially available from Avanti Polar Lipids, USA) were dissolved in chloroform and the final lipid concentration was 5 mg/mL. The POPC/b-PE solution was vortex dried under a slow stream of nitrogen to form a thin, uniform POPC/b-PE film. The POPC/b-PE film was further dried in a vacuum chamber overnight to remove residual chloroform.
    • (2) The POPC/biotin-PE film in step (1) was dispersed in and mixed with a phosphate buffer containing 10 mM of phosphate buffered saline, 150 mM of sodium chloride aqueous solution, and 0.02% (w/v) of sodium azide (NaN3, commercially available from Sigma-Aldrich, USA), with the pH adjusted to 7.2, The mixed solution was filtered through the 100-nm, followed by the 50-nm Nuclepore® track-etched polycarbonate membranes (Whatman Schleicher & Schnell, Germany) at least 10 times under 150 psi at room temp.
    • (3) The filtered solution in step (2) was passed through the LIPEX™ Extruder (Northern Lipids, Inc. Canada) to generate a homogenous population of unilamillar vesicles. The size of the POPC/biotin-PE vesicles was about 65±3 nm, determined by the dynamic laser light scattering detector (Zetasizer Nano ZS, Malvern Instruments, Germany).
    Preparation of the Bioactive Composition
  • Biotinylated EpCAM Antibody was prepared by the following steps:
    • (1) The anti-EpCAM monoclonal antibody (OC98-1 or EpAb4-1) was generated by method described by Chen et al (Clip Vaccine Immunol 2007; 14:404-11).
    • (2) The antibody in step (1) was dissolved in a buffer solution containing 10 mM of PBS and 150 mM of NaCl, with a pH about 7.2, The concentration of the antibody buffer solution was about 0.65 mg/mL, determined by Nanodrop 1000 spectrophotometer (Thermo Scientific, USA).
    • (3) The antibody solution in step (2) was mixed with 10 mM of Sulfo NHS-LC-Biotin (with a molar ratio of 1 to 10) and dissolved in Milli-Q water (Milli-Q RO system, USA) at room temperature for 30 min. Excess biotin was removed by dialysis in phosphate buffered saline at 4° C. for 24 h, with a buffer change every 12 h.
    • (4) The ratio of biotin and antibody in the biotinylated anti-EpCAM antibody (bOC98-1 or bEpAb4-1) was 1.5 to 1, determined by the HABA assay using a biotin quantitation kit (Pierce, USA).
  • Alternatively, commercially available biotinylated goat anti-human anti-EpCAM antibody from R and D Systems (Minneapolis, Minn.) could be used.
  • Preparation of Solid Substrates of the Present Invention
  • Glass substrate (such as microscope coverslips from Deckglaser, Germany) were cleaned with 10% DECON 90 (Devon Laboratories Limited, England), rinsed with Milli-Q water, dried under nitrogen gas, and exposed to oxygen plasma in a plasma cleaner (Harrick Plasma, Ithaca, N.Y., U.S.A.) at 100 mtorr for 10 min. Prior to each use, the glass substrate was rinsed with ethanol and dried under nitrogen gas.
  • Silicon oxide based solid substrates (e.g. silicon wafer or glass coverslips) were cleaned with piranha solution (70% sulfuric acid and 30% hydrogen peroxide (v/v)) at 120° C. tor 40 min, subsequenctly washed with distilled water and rinsed with acetone. The solid substrates were dried under a stream of nitrogen and treated with a plasma cleaner.
  • For the vapor phase silanization reaction, clean silicon oxide substrates and a Petri-dish containing 150 μL of 3-(aminopropyl)-triethoxysilane (Sigma, USA) were placed in a desiccator (Wheaton dry-seal desiccator, 100 nm) under reduced pressure at ˜0.3 Torr for 16 h. The substrates were cleaned by acetone and dried under nitrogen stream.
  • Construction of the SLB Surface Coating on a Solid Substrate
  • 0.25 mg/ml of POPC/b-PE vesicle solution from paragraph [0084] was added to the cleaned solid substrate to form a SLB coated solid substrate. This was followed by an extensive rinse with a phosphate buffer containing 10 mM PBS and 150 mM NaCl (pH=7.2) to remove excess POPC/b-PE vesicles. Biotin was the functional group in the SLB which binds with the functional group (streptavidin) in the linker composition
  • 0.1 mg/mL of streptavidin (SA) solution (commercially available from Pierce Biotechnology, Rockford, Ill., USA) was added to the SLB coated solid substrate and incubated for 1 hour, followed with a PBS buffer rinse to remove excess SA.
  • About 0.05 mg/mL of b-Anti-EpCAM solution was added to the SA-SLB coated solid substrate to form the surface coating of the present invention.
  • Construction of the PEG Surface Coating on a Solid Substrate
  • The biotinylated PEG si lane solution (Si-bPEGs) was added to the clean glass substrate and incubated for 1 hour to form a Si-bPEG nonfouling composition on the glass substrate, followed by an ethanol rinse to remove excess Si-bPEGs. Silane was the surface linker and the biotin was the functional group that bind with the functional group (SA) in the linker composition.
  • 0.1 mg/mL of SA solution was added to the Si-bPEGs coated solid substrate and incubated for 1 hour, followed by a PBS buffer rinse to remove excess SA.
  • 0.05 mg/mL of b-Anti-EpCAM solution was added and bound with SA-Si-bPEGs surface coating, followed by PBS buffer rinse to remove excess b-Anti-EpCAM.
  • Construction of the PENT Surface Coating on a Solid Substrate
  • Physical deposition of PEM films was performed by batch and static conditions as follows: initially, all polypeptides were dissolved in 10 mM Tris-HCl buffer with 0.15 M NaCl, pH 7.4. Solid substrates were then immersed in PLL (MW 15000-30000; Sigma, St Louis, Mo.) solution (1 mg/mL) for 10 min at room temperature, followed by rinsing with 1 mL of Tris-HCl buffer for 1 min. To couple PLGA, the PLL-coated slide was subsequently immersed in the PLGA solution (MW 3000-15000, Sigma, St Louis, Mo., 1 mg/mL) for 10 min, followed by rinsing with 1 mL of Tris-HCl buffer for 1 min. Lastly, substrates were cleaned with fresh PBS to remove uncoupled polypeptides. The resulting c-(PLL/PLGA)i, where i was denoted as the number of polyelectrolyte pairs generated by repeating the above steps: i) 0.5 was referred to c-PLL only, i) I was referred to c-(PLL/PLGA)1, and the like.
  • QCM-D Characterization of the SLB Surface Coating
  • The construction of the surface coating was monitored by quartz crystal microbalance with dissipation (QCM-D). The QCM-D response in FIG. 11 shows the construction of the surface coating on a SiO2-pretreated quartz crystal. First, 0.25 mg/mL of POPC/b-PE vesicle mixture (in phosphate buffer) was dispensed into the QCM chamber at point (I). The normalized frequency change F and dissipation shift D were 26.0±0.7 Hz and 0.19±0.03×10-6 respectively, which are the characteristics of a highly uniformed lipid bilayer. After two buffer washes (denoted as *), 0.1 mg/mL, of SA solution was dispensed at point II. • SA binding was saturated at F=52.8±5.4 Hz and D=3.84±0.54×10-6. At point (III), 0.025 mg/mL of OC98-1 antibody solution was dispensed into the QCM chamber and there was no frequency or dissipation change. This shows there was no interaction between the OC98-1 antibody and the SA-lipid bilayer surface. In contrast, adding biotinylated antibody solution (OC98-1 or bEpAb4-1) at point (IV) resulted in frequency and dissipation change, with equilibrated shifts of F=39.4±6.8 Hz and D=1.63±0.28×10-6. This demonstrates the binding of biotinylated antibody to SA-lipid bilayer surface.
  • The characteristics of the SLB nonfouling composition on the surface coating were examined using QCM-D (FIG. 12). Bovine serum albumin (BSA, commercially available from Sigma-Aldrich, USA) was added to the surface coating and there was a sudden change in frequency and dissipation, with equilibrated shifts of F=6.9 Hz and D=3.35×10-6. This indicates an immediate BSA adsorption. Three buffer rinses (*) caused an increase in frequency and a decrease in disspation, with saturated shifts of F=6.1 Hz and D=3.16×10-6. This indicates the adsorbed BSA can be easily removed from the surface coating and thus, a very weak interaction between BSA and SLB.
  • Example 2 Preparation of the Microfluidic Chip
  • The microfluidic p can be prepared by the following steps:
    • 1. A commercial CO2 laser scriber (Helix 24, Epilog, USA) was used to engrave the microtrenches to form microstructures on the PMMA substrate.
    • 2. The PMMA substrate, glass substrate and nuts were cleaned with MeOH, detergent and water, followed by 10 min sonication. The nuts and the solid substrates were dried by nitrogen gas and baked for 10 min at 60° C.
    • 3. The PMMA substrate gas bon led with nuts by chloroform treatment.
    • 4. PMMA substrate and the glass slide were joined together using an adhesive (e.g. 3M doubled sided tape from 3M, USA).
    Example 3 CTCs Binding to the Anti-EpCAM Functionalized SLB Surface Coating
  • Eight blood samples were used to determine the CTC capture rate of the Anti-EpCAM functionalized SLB surface coating in a microfluidic chip in Example 2. Each blood sample contained 2 ml of blood from a stage IV colon cancer patient and the sample was introduced to the sealed channel of the microfluidic chip at 0.5 ml/hr, controlled by a syringe pump. Subsequently, the sealed channel in the microfluidic chip was rinsed with 0.5 ml of PBS buffer at the flow rate of 1 ml/h, followed by in situ immunostaining.
  • The number of CTCs captured per ml of blood for these 8 samples were 26, 34, 36, 39, 47, 67 79, and 99. 25% of the blood samples had 79 or higher CTC count per ml of testing sample and the median CTC count was 43 per ml of testing sample. There was minimal binding of the non-specific cells and proteins after the buffer rinse.
  • As a comparison, the CTC count for the FDA approved Veridex CellSearch is as follows: 25% of the samples had 3 or more CTCs per 7.5 ml of testing sample and the median CTC counts was 0.
  • The anti-EpCAM functionalized SLB surface was incubated with 150 uL of HCT116 cancer cell spiked human blood (with HCT116 cancer cell density of approximately 10 to 100 per 100 μL of blood), followed by a buffer rinse to remove non-specific cells. FIG. 13 shows the surface coating before and after the buffer rinse. Prior to the buffer rinse, the surface coating was covered with non-specific cells (upper left) and four HCT116 cancer cells (lower left). After the buffer rinse, almost all of the non-specific cells were removed (upper right) but the four HCT116 cancer cell (lower right) remained on the surface coating.
  • The results show the surface coating of the present invention effective in capturing CTCs and releasing the non-specific cells.
  • Example 4 Comparison of Capture Efficiency and Nonfouling Property of Various Surface Conditions
  • The capture rate of HCT116 cancer cells (biological substance) and the nonfouling property of six different surface conditions are illustrated in FIG. 14A,
  • The results show that the surface coatings of the present invention (lipid/SA/b-anti-EpCAM and PEG(15 mM)/SA/b-anti-EpCAM) are more effective in capturing the biological substance. There is less binding of the non-specific cells (white blood cells or WBC) on the surface coatings of the present invention compare to a surface coating without a nonfouling composition (glass only).
  • FIG. 14B shows the non-specific, blood cell binding of the following surfaces: (A) Glass only; (B) biotinylated SLB (b-SLB), (C) Streptavidin conjugated-bSLB, and (D) OC98-1-conjugated bSLB. These surfaces were incubated with diluted human blood from healthy donor (1 uL, of blood in 100 uL PBS buffer) for 4 hours, followed by a PBS buffer rinse. Images (E) to (H) are the after rinse images which correspond to the surface coatings in (A) to (D). The results show that after a buffer rinse, there is less non-specific blood cell on the surface coatings with a releasable composition (i.e. SLB) compare to the surface coating without a releasable composition (i.e. glass only).
  • Example 5 Purification by Flow
  • The differentiated flow shear could selectively “flush” out the non-specific cells based on the affinity of these cells to the nonfouling composition, while the biological substance remains on the surface coating.
  • In this study, the surface coating comprised a SLB, a linker composition and fibronectin as the bioactive composition. FIG. 15A shows fibroblast 3T3 (green) and colon cancer cell line HCT116 (red) were incubated on the surface coating for 4 h. The surface coating was rinsed with a buffer solution, which has a shear stress of 3 dyne/cm2.
  • The HCT 116 cells (red) were flushed away from the surface coating within 5 min of the buffer rinse, as shown in FIG. 15B. The fibroblast 3T3 cells (green) remained on the surface coating after 30min of buffer rinse, as shown in FIG. 15C, due to its high affinity to fibronectin.
  • The result shows a shear stress about 3 dyne/cm2 is sufficient to remove the non-specific cells from the releasable composition.
  • FIG. 16 summarizes the respective shear stress and flushing time for the HCT116 and NIH-3T3 cell populations (non-specific cells). To remove HCT116 cells from the releasable composition of the surface coating, the shear stress is about 3 to about 4.5 dyne/cm2. To remove NIH-3T3 cells from the releasable composition of the surface coating, the shear stress is about 8.5 to about 12 dyne/cm2 (N/N0 is the percentage of the cells remains attached to the surface coating using various shear stresses, N is the final cell number and N0 is the initial cell number.)
  • Example 6 Release of CTCs from the Surface Coating
  • The captured HCT116 cancer cells on the surface coating in Example 3 were released by introducing air bubbles. FIG. 17 shows HCT116 cells in the red circle were removed from the surface coating within 3 seconds of introducing air bubbles.
  • Example 7 Culture of Released CTCs From the Surface Coating
  • The captured CTCs were incubated with 5 mM of EDTA at 37° C. for 5 to 10 min and released by flowing a culture medium into the sealed channel of the microfluidic chip. A total of 18 colo205 cells were released from this procedure. The released colo205 cells, together with a serum-containing culture medium and antibiotics (penicillin+streptomycin+gentamicin), were placed into a 48-well tissue cultured polystyrene plate for cultivation.
  • FIGS. 18A-18C show a portion of 18 colo205 cells on day 1, on day 10 and day 14 respectively. This study demonstrates the released colo205 cells retained their viability for subsequent cell culture.
  • Example 8 Capture CTCs Through a CTC Filtration Device
  • Any membranes, tubes, capillaries, beads, nanoparticles or channels can be coated with the surface coating of the present invention. FIG. 19 illustrates schematically a filtration device, wherein the filtration filter is coated with the surface coating of the present invention. The filter could accommodate high volume blood flow and capture a biological substance for a diagnostic or therapeutic purpose. To access the patient's blood or body fluid, a catheter can be inserted into the patient's vein or fistula and the patient's blood flows through the CTC filtration device, wherein the surface coating on the filters captures the CTCs. The filtered blood flows back to the patient.
  • Example 9 Capture CTCs Through a Biotinylated EpAb4-1 Antibody
  • The binding specificity of biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody and biotinylated EpCam antibody (commercially available from R&D system, USA) were examined using the HCT116 (colorectal) CTCs and SAS (tongue) CTCs.
  • The CTCs were spiked in a buffer solution (about 105 CTCs/ ml). The CTC-spiked buffer solution was introduced to the surface coatings with the following bioactive composition: biotinylated OC9801 antibody, biotinylated EpAb4-1 antibody, biotinylated EpCam antibody and IgC1 antibody.
  • The CTC binding specificy of the antibodies was determined by colorimetric method, by measuring the absorption optical density at 490 nm. FIG. 20 shows biotinylated EpAb 4-1 is effective in capturing HCT116 CTCs and SAS CTCs.

Claims (21)

1.-63. (canceled)
64. A microfluidic chip for selectively enriching rare cells, comprising:
a first solid substrate and a second solid substrate, wherein at least one of the first and second solid substrates comprise a series of microstructures configured to interact with cells, and wherein the first and second solid substrates are configured to be bound parallel to one another; and
a surface coating for capturing the rare cells, wherein the surface coating comprises a non-fouling composition and a bioactive composition which selectively binds to the rare cells, wherein the non-fouling composition of the surface coating is non-covalently associated with the bioactive composition,
wherein each of the first and second solid substrates comprise the surface coating.
65. The microfluidic chip of claim 64, wherein the microstructures are ordered such that progressing from one side of the microfluidic chip to the other side of the microfluidic chip longitudinally, the openings between the microstructures are staggered.
66. The microfluidic chip of claim 64, wherein the bioactive composition comprises an antibody.
67. The microfluidic chip of claim 66, wherein the antibody is a biotinylated EpCAM antibody.
68. The microfluidic chip of claim 64, wherein the non-fouling composition comprises a lipid layer.
69. The microfluidic chip of claim 64, wherein the two solid substrates comprises a glass substrate and a plastic substrate.
70. The microfluidic chip of claim 69, wherein the glass substrate is located below the plastic substrate in a working configuration.
71. The microfluidic chip of claim 64, wherein the first solid substrate comprises the series of microstructures, and wherein the first solid substrate is located above the second solid substrate in a working configuration.
72. The microfluidic chip of claim 64, further comprising an adhesive for bonding the first solid substrate to the second solid substrate.
73. The microfluidic chip of claim 72, wherein the adhesive comprises an inner hollow opening in a form of a channel.
74. The microfluidic chip of claim 73, wherein the channel is configured to encompass the series of microstructures.
75. The microfluidic chip of claim 73, wherein the channel of the adhesive determines a path for the rare cells to travel through for the microfluidic chip.
76. The microfluidic chip of claim 72, wherein a thickness of the adhesive determines a height of a channel of the microfluidic chip.
77. The microfluidic chip of claim 64, wherein the binding moiety comprises an antibody, and the antibody comprises a heavy chain and a light chain that binds EpCAM, wherein (a) the heavy chain comprises CDR1, CDR2, and CDR3 of SEQ ID No: 1, and (b) the light chain comprises CDR1, CDR2, and CDR3 of SEQ ID NO: 2.
78. The microfluidic chip of claim 64, further comprising a syringe pump wherein the syringe pump is configured to apply buffer at a flow rate configured to release non-specific cells from the non-fouling layer without releasing cells selectively bound to the bioactive composition.
79. The microfluidic chip of claim 64, further comprising a syringe pump configured to aid rinsing the microfluidic chip with a buffer at a shear force of about 2.5 to about 10 dyne/cm2.
80. The microfluidic chip of claim 64, wherein the non-fouling composition is coupled to each of the first and second solid substrates by a surface linker.
81. The microfluidic chip of claim 64, wherein the non-fouling composition is from 2 nm to 300 um thick.
82. The microfluidic chip in accordance with claim 64, wherein the surface coating is attached to the solid substrate by one of the following non-covalent interactions: covalent bonding, hydrogen bonding, electrostatic interaction, hydrophilic-hydrophilic interaction, polar-polar interaction, magnetic force, or a combination thereof.
83. The microfluidic chip of claim 64, wherein the non-fouling composition is configured to completely coat each of the first and second solid substrates.
US15/378,938 2011-06-29 2016-12-14 Capture, purification, and release of biological substances using a surface coating Abandoned US20170199184A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/378,938 US20170199184A1 (en) 2011-06-29 2016-12-14 Capture, purification, and release of biological substances using a surface coating
US16/832,396 US11674958B2 (en) 2011-06-29 2020-03-27 Capture, purification, and release of biological substances using a surface coating
US18/308,531 US20230366879A1 (en) 2011-06-29 2023-04-27 Capture, purification, and release of biological substances using a surface coating

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161502844P 2011-06-29 2011-06-29
US201261606220P 2012-03-02 2012-03-02
PCT/US2012/044701 WO2013003624A2 (en) 2011-06-29 2012-06-28 The capture, purification and release of biological substance using a surface coating
US201414128354A 2014-05-20 2014-05-20
US15/378,938 US20170199184A1 (en) 2011-06-29 2016-12-14 Capture, purification, and release of biological substances using a surface coating

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2012/044701 Continuation WO2013003624A2 (en) 2011-06-29 2012-06-28 The capture, purification and release of biological substance using a surface coating
US14/128,354 Continuation US9541480B2 (en) 2011-06-29 2012-06-28 Capture, purification, and release of biological substances using a surface coating

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/832,396 Continuation US11674958B2 (en) 2011-06-29 2020-03-27 Capture, purification, and release of biological substances using a surface coating

Publications (1)

Publication Number Publication Date
US20170199184A1 true US20170199184A1 (en) 2017-07-13

Family

ID=47424802

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/128,354 Active 2033-05-02 US9541480B2 (en) 2011-06-29 2012-06-28 Capture, purification, and release of biological substances using a surface coating
US15/378,938 Abandoned US20170199184A1 (en) 2011-06-29 2016-12-14 Capture, purification, and release of biological substances using a surface coating
US16/832,396 Active US11674958B2 (en) 2011-06-29 2020-03-27 Capture, purification, and release of biological substances using a surface coating
US18/308,531 Pending US20230366879A1 (en) 2011-06-29 2023-04-27 Capture, purification, and release of biological substances using a surface coating

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/128,354 Active 2033-05-02 US9541480B2 (en) 2011-06-29 2012-06-28 Capture, purification, and release of biological substances using a surface coating

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/832,396 Active US11674958B2 (en) 2011-06-29 2020-03-27 Capture, purification, and release of biological substances using a surface coating
US18/308,531 Pending US20230366879A1 (en) 2011-06-29 2023-04-27 Capture, purification, and release of biological substances using a surface coating

Country Status (5)

Country Link
US (4) US9541480B2 (en)
EP (1) EP2726870B1 (en)
CN (3) CN110763842A (en)
TW (2) TWI550273B (en)
WO (1) WO2013003624A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane
US10112198B2 (en) 2014-08-26 2018-10-30 Academia Sinica Collector architecture layout design
WO2019025437A1 (en) 2017-07-31 2019-02-07 Universidad De Cantabria Cartridge, device and method for detecting, capturing, identifying and counting circulating tumour cells
US10495644B2 (en) 2014-04-01 2019-12-03 Academia Sinica Methods and systems for cancer diagnosis and prognosis
US11674958B2 (en) 2011-06-29 2023-06-13 Academia Sinica Capture, purification, and release of biological substances using a surface coating

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2848506C (en) 2011-09-14 2020-07-21 Neupharma, Inc. Certain chemical entities, compositions, and methods
WO2013173497A1 (en) * 2012-05-15 2013-11-21 Massachusetts Institute Of Technology Systems and methods for detecting molecular interactions using magnetic beads
US9494500B2 (en) 2012-10-29 2016-11-15 Academia Sinica Collection and concentration system for biologic substance of interest and use thereof
WO2016019401A1 (en) * 2014-07-30 2016-02-04 Medvisionus Llc Microfluidic device with smooth surface for enrichment of rare cells and biomarkers from a biological fluid
WO2016086374A1 (en) * 2014-12-03 2016-06-09 士捷医疗设备(武汉)有限公司 Microfluidic device for simulating environment in cell body and application thereof
US20160178490A1 (en) 2014-12-22 2016-06-23 Saint-Gobain Performance Plastics Corporation Capture system of cells and methods
US10280390B2 (en) 2014-12-22 2019-05-07 Saint-Gobain Performance Plastics Corporation System for culture of cells in a controlled environment
US11175286B2 (en) 2015-01-09 2021-11-16 Spot Biosystems Ltd. Immunolipoplex nanoparticle biochip containing molecular probes for capture and characterization of extracellular vesicles
CN106148315B (en) * 2015-04-14 2019-02-01 中国科学院苏州纳米技术与纳米仿生研究所 A kind of CTC capture based on chitin nanometer and purifying substrate and preparation method thereof
KR102384741B1 (en) 2015-04-22 2022-04-07 버클리 라잇츠, 인크. Microfluidic Cell Culture
ES2865259T3 (en) * 2015-07-20 2021-10-15 Sentilus Holdco LLC Chips, detectors and methods to make and use them
US10799865B2 (en) 2015-10-27 2020-10-13 Berkeley Lights, Inc. Microfluidic apparatus having an optimized electrowetting surface and related systems and methods
EP3387438B1 (en) 2015-12-08 2023-03-01 Berkeley Lights, Inc. Microfluidic devices and kits and methods for use thereof
CN108806872A (en) * 2016-03-20 2018-11-13 李忠波 A kind of mould proof fungi-proofing anticorrosive cable of transmission of electricity
CN107287107A (en) * 2016-03-30 2017-10-24 无锡纳奥生物医药有限公司 A kind of circulating tumor cell separation equipment, system and method
SG10202008265XA (en) * 2016-05-26 2020-09-29 Berkeley Lights Inc Covalently modified surfaces, kits, and methods of preparation and use
CN107589254B (en) * 2016-07-08 2022-02-08 鉴识生物系统有限公司 Preparation method and application of immunoliposome complex nanoparticle biochip
US11085071B2 (en) 2017-02-01 2021-08-10 Spot Biosystems Ltd. Highly stable and specific molecular beacons encapsulated in cationic lipoplex nanoparticles and application thereof
US9738937B1 (en) 2017-03-31 2017-08-22 Cellmax, Ltd. Identifying candidate cells using image analysis
CN109097243A (en) * 2017-06-21 2018-12-28 曦医生技股份有限公司 Biological particle captures chipset
CN107570482A (en) * 2017-07-06 2018-01-12 天津大学 The removal device and method of the non-specific adsorption thing at interface
TWI642685B (en) * 2017-07-26 2018-12-01 中原大學 Novel peptide and application thereof
US11709156B2 (en) 2017-09-18 2023-07-25 Waters Technologies Corporation Use of vapor deposition coated flow paths for improved analytical analysis
US11709155B2 (en) 2017-09-18 2023-07-25 Waters Technologies Corporation Use of vapor deposition coated flow paths for improved chromatography of metal interacting analytes
CN107936085A (en) * 2017-11-21 2018-04-20 西南交通大学 Using ultraviolet irradiation in TiO2The method and TiO of upper ankyrin and regulating cell compatibility2Protein product
CN111512158B (en) * 2017-12-22 2023-12-12 威斯康星校友研究基金会 Nanoengineered surfaces for cancer biomarker capture
WO2019161165A1 (en) 2018-02-15 2019-08-22 Ohio State Innovation Foundation Microfluidic devices and methods for high throughput electroporation
CN108795692B (en) * 2018-06-26 2021-01-01 成都普瑞康生物科技有限公司 Rare cell capture system and application thereof
CN109187943B (en) * 2018-08-24 2021-07-30 四川新健康成生物股份有限公司 Anti-interference reagent cup and preparation method of anti-interference coating in reagent cup
CN109136087A (en) * 2018-09-11 2019-01-04 京东方科技集团股份有限公司 Separating chips and separation method
CN109880148B (en) * 2019-01-22 2021-08-06 肇庆医学高等专科学校 Preparation of surface imprinting material and application of surface imprinting material in glutamic acid enantiomer resolution
CN110133267A (en) * 2019-06-18 2019-08-16 温州医科大学附属第二医院、温州医科大学附属育英儿童医院 It is a kind of for adsorbing the carrier of tumour cell, preparation method, kit and application
CN112300994B (en) * 2019-08-02 2024-01-12 中国科学院苏州纳米技术与纳米仿生研究所 Nanometer magnetic bead for capturing circulating tumor cells and preparation method and application thereof
CN115812104A (en) * 2020-03-27 2023-03-17 加利福尼亚大学董事会 Covalent chemistry allows purification of extracellular vesicles on nano-substrates-early detection of hepatocellular carcinoma
CN111472200B (en) * 2020-05-19 2021-12-24 太原理工大学 Method for in-situ construction of anti-fouling gel coating on surface of cellulose test paper
CN111632202A (en) * 2020-06-29 2020-09-08 南开大学 Tumor cell adhesion material and preparation method and application thereof
CN112501122A (en) * 2020-12-09 2021-03-16 河南大学 Method for separating and extracting tumor cells
CN112646635A (en) * 2020-12-22 2021-04-13 谢强 Lubricating oil composition and preparation method thereof
WO2022184093A1 (en) * 2021-03-03 2022-09-09 华联生物科技股份有限公司 Bioelectronic system for rare cell separation and application thereof

Family Cites Families (289)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3784015A (en) 1972-02-17 1974-01-08 Bendix Corp Filter
US5147606A (en) 1990-08-06 1992-09-15 Miles Inc. Self-metering fluid analysis device
US5646001A (en) 1991-03-25 1997-07-08 Immunivest Corporation Affinity-binding separation and release of one or more selected subset of biological entities from a mixed population thereof
US5652148A (en) 1993-04-20 1997-07-29 Actimed Laboratories, Inc. Method and apparatus for red blood cell separation
US5427663A (en) 1993-06-08 1995-06-27 British Technology Group Usa Inc. Microlithographic array for macromolecule and cell fractionation
US5554686A (en) 1993-08-20 1996-09-10 Minnesota Mining And Manufacturing Company Room temperature curable silane-terminated polyurethane dispersions
US5707799A (en) 1994-09-30 1998-01-13 Abbott Laboratories Devices and methods utilizing arrays of structures for analyte capture
US6718053B1 (en) 1996-11-27 2004-04-06 Chromavision Medical Systems, Inc. Method and apparatus for automated image analysis of biological specimens
ATE236386T1 (en) 1995-11-30 2003-04-15 Chromavision Med Sys Inc METHOD FOR AUTOMATIC IMAGE ANALYSIS OF BIOLOGICAL SAMPLES
US5885470A (en) 1997-04-14 1999-03-23 Caliper Technologies Corporation Controlled fluid transport in microfabricated polymeric substrates
US6790366B2 (en) 1996-06-07 2004-09-14 Immunivest Corporation Magnetic separation apparatus and methods
US6890426B2 (en) 1996-06-07 2005-05-10 Immunivest Corporation Magnetic separation apparatus and methods
US6074827A (en) 1996-07-30 2000-06-13 Aclara Biosciences, Inc. Microfluidic method for nucleic acid purification and processing
EP0929658A4 (en) 1996-08-26 2005-11-02 Univ Princeton Reversibly sealable microstructure sorting devices
US6039897A (en) 1996-08-28 2000-03-21 University Of Washington Multiple patterned structures on a single substrate fabricated by elastomeric micro-molding techniques
WO1998023948A1 (en) * 1996-11-29 1998-06-04 The Board Of Trustees Of The Leland Stanford Junior University Arrays of independently-addressable supported fluid bilayer membranes and methods of use thereof
US6583251B1 (en) 1997-09-08 2003-06-24 Emory University Modular cytomimetic biomaterials, transport studies, preparation and utilization thereof
US5842787A (en) 1997-10-09 1998-12-01 Caliper Technologies Corporation Microfluidic systems incorporating varied channel dimensions
JP2001520236A (en) 1997-10-22 2001-10-30 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Spacer peptide and membrane containing it
CA2320418C (en) 1998-02-12 2008-06-17 Leon W.M.M. Terstappen Methods and reagents for the rapid and efficient isolation of circulating cancer cells
US7282350B2 (en) 1998-02-12 2007-10-16 Immunivest Corporation Labeled cell sets for use as functional controls in rare cell detection assays
SE521415C2 (en) 1998-02-17 2003-10-28 Hans Goeran Evald Martin Method for producing a gas sensor-associated detector, as well as a detector made according to the method
WO1999052574A1 (en) 1998-04-10 1999-10-21 Massachusetts Institute Of Technology Biopolymers resistant coatings
US6361749B1 (en) 1998-08-18 2002-03-26 Immunivest Corporation Apparatus and methods for magnetic separation
US20010036556A1 (en) 1998-10-20 2001-11-01 James S. Jen Coatings for biomedical devices
WO2000047998A1 (en) 1999-02-10 2000-08-17 Cell Works Inc. Class characterization of circulating cancer cells isolated from body fluids and methods of use
US6153113A (en) 1999-02-22 2000-11-28 Cobe Laboratories, Inc. Method for using ligands in particle separation
US6322683B1 (en) 1999-04-14 2001-11-27 Caliper Technologies Corp. Alignment of multicomponent microfabricated structures
WO2000078920A1 (en) 1999-06-21 2000-12-28 The General Hospital Corporation Methods and devices for cell culturing and organ assist systems
US6623982B1 (en) 1999-07-12 2003-09-23 Immunivest Corporation Increased separation efficiency via controlled aggregation of magnetic nanoparticles
US6790599B1 (en) 1999-07-15 2004-09-14 Microbionics, Inc. Microfluidic devices and manufacture thereof
US6271309B1 (en) 1999-07-30 2001-08-07 3M Innovative Properties Company Curable compositions comprising the hydrosilation product of olefin-containing polymers and organosiloxane hydrides, cured compositions made therefrom, and methods of making same
ATE386747T1 (en) 1999-12-27 2008-03-15 Crucell Holland Bv ANTIBODIES AGAINST EP-CAM
DE60142220D1 (en) 2000-02-16 2010-07-08 Wisconsin Alumni Res Found BIOCHEMICAL BLOCKING LAYER FOR LIQUID CRYSTAL
WO2001071027A2 (en) 2000-03-24 2001-09-27 Micromet Ag mRNA AMPLIFICATION
AU2001260153B2 (en) 2000-03-24 2006-08-17 Micromet Ag Multifunctional polypeptides comprising a binding site to an epitope of the NKG2D receptor complex
US7769420B2 (en) 2000-05-15 2010-08-03 Silver James H Sensors for detecting substances indicative of stroke, ischemia, or myocardial infarction
US20020182633A1 (en) 2000-07-11 2002-12-05 Chen Christopher S. Methods of patterning protein and cell adhesivity
AU8843701A (en) 2000-09-09 2002-03-22 Univ New York State Res Found Method and compositions for isolating metastatic cancer cells, and use in measuring metastatic potential of a cancer thereof
NL1016779C2 (en) 2000-12-02 2002-06-04 Cornelis Johannes Maria V Rijn Mold, method for manufacturing precision products with the aid of a mold, as well as precision products, in particular microsieves and membrane filters, manufactured with such a mold.
US6777836B2 (en) 2000-12-20 2004-08-17 General Electric Company Heat transfer enhancement at generator stator core space blocks
US6685841B2 (en) 2001-02-14 2004-02-03 Gabriel P. Lopez Nanostructured devices for separation and analysis
US6913697B2 (en) 2001-02-14 2005-07-05 Science & Technology Corporation @ Unm Nanostructured separation and analysis devices for biological membranes
US20060014013A1 (en) 2001-03-10 2006-01-19 Saavedra Steven S Stabilized biocompatible supported lipid membrane
EP1385692B1 (en) 2001-04-06 2011-03-02 Fluidigm Corporation Polymer surface modification
PL205352B1 (en) * 2001-05-03 2010-04-30 Merck Patent Gmbh Recombinant tumor specific antibody and use thereof
US6844028B2 (en) 2001-06-26 2005-01-18 Accelr8 Technology Corporation Functional surface coating
US7501157B2 (en) 2001-06-26 2009-03-10 Accelr8 Technology Corporation Hydroxyl functional surface coating
US7858679B2 (en) 2001-07-20 2010-12-28 Northwestern University Polymeric compositions and related methods of use
WO2003008376A2 (en) 2001-07-20 2003-01-30 Northwestern University Adhesive dopa-containing polymers and related methods of use
US8815793B2 (en) 2001-07-20 2014-08-26 Northwestern University Polymeric compositions and related methods of use
US7863012B2 (en) 2004-02-17 2011-01-04 Veridex, Llc Analysis of circulating tumor cells, fragments, and debris
US20050230272A1 (en) 2001-10-03 2005-10-20 Lee Gil U Porous biosensing device
WO2003052420A2 (en) 2001-10-03 2003-06-26 Purdue Research Foundatio Device and bioanalytical method utilizing asymmetric biofunction alized membrane
US6949355B2 (en) 2001-10-11 2005-09-27 Aviva Biosciences Methods, compositions, and automated systems for separating rare cells from fluid samples
US8980568B2 (en) 2001-10-11 2015-03-17 Aviva Biosciences Corporation Methods and compositions for detecting non-hematopoietic cells from a blood sample
US8986944B2 (en) 2001-10-11 2015-03-24 Aviva Biosciences Corporation Methods and compositions for separating rare cells from fluid samples
EP1448807A4 (en) 2001-10-30 2005-07-13 Massachusetts Inst Technology Fluorocarbon-organosilicon copolymers and coatings prepared by hot-filament chemical vapor deposition
US20030163084A1 (en) 2001-12-20 2003-08-28 Klaus Tiemann Creation and agitation of multi-component fluids in injection systems
AU2003216175A1 (en) 2002-02-04 2003-09-02 Colorado School Of Mines Laminar flow-based separations of colloidal and cellular particles
GB0207350D0 (en) 2002-03-28 2002-05-08 Univ Sheffield Surface
CN1646912A (en) * 2002-04-03 2005-07-27 独立行政法人科学技术振兴机构 Biochip sensor surface carrying polyethylene glycolated nanoparticles
US6955738B2 (en) 2002-04-09 2005-10-18 Gyros Ab Microfluidic devices with new inner surfaces
US7217562B2 (en) 2002-04-16 2007-05-15 Princeton University Gradient structures interfacing microfluidics and nanofluidics, methods for fabrication and uses thereof
US7141369B2 (en) 2002-04-25 2006-11-28 Semibio Technology, Inc. Measuring cellular metabolism of immobilized cells
SE0201738D0 (en) 2002-06-07 2002-06-07 Aamic Ab Micro-fluid structures
US7696320B2 (en) 2004-08-24 2010-04-13 Domantis Limited Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
US8911831B2 (en) 2002-07-19 2014-12-16 Northwestern University Surface independent, surface-modifying, multifunctional coatings and applications thereof
WO2004011669A2 (en) 2002-07-30 2004-02-05 University Of Washington Apparatus and methods for binding molecules and cells
US20040109853A1 (en) 2002-09-09 2004-06-10 Reactive Surfaces, Ltd. Biological active coating components, coatings, and coated surfaces
US20100210745A1 (en) 2002-09-09 2010-08-19 Reactive Surfaces, Ltd. Molecular Healing of Polymeric Materials, Coatings, Plastics, Elastomers, Composites, Laminates, Adhesives, and Sealants by Active Enzymes
US20110250626A1 (en) 2002-09-09 2011-10-13 Reactive Surfaces, Ltd. Visual Assays for Coatings Incorporating Bioactive Enzymes for Catalytic Functions
US20100233146A1 (en) 2002-09-09 2010-09-16 Reactive Surfaces, Ltd. Coatings and Surface Treatments Having Active Enzymes and Peptides
US20110240064A1 (en) 2002-09-09 2011-10-06 Reactive Surfaces, Ltd. Polymeric Coatings Incorporating Bioactive Enzymes for Cleaning a Surface
ES2375724T3 (en) 2002-09-27 2012-03-05 The General Hospital Corporation MICROFLUDE DEVICE FOR SEPERATION OF CELLS AND ITS USES.
US8129330B2 (en) 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
DE10246069A1 (en) 2002-10-02 2004-04-15 Robert Bosch Gmbh Device for determining at least one parameter of a medium flowing in a line
ES2341103T3 (en) 2002-10-23 2010-06-15 The Trustees Of Princeton University CONTINUOUS SEPARATION PROCEDURE OF PARTICLES USING RETICULATES OF ASYMETRICALLY ALIGNED OBSTACLES REGARDING FIELDS.
CA2518667C (en) 2003-03-14 2011-07-19 The Trustees Of Columbia University In The City Of New York Systems and methods of blood-based therapies having a microfluidic membraneless exchange device
US20060076295A1 (en) 2004-03-15 2006-04-13 The Trustees Of Columbia University In The City Of New York Systems and methods of blood-based therapies having a microfluidic membraneless exchange device
US20070010702A1 (en) 2003-04-08 2007-01-11 Xingwu Wang Medical device with low magnetic susceptibility
US20050107870A1 (en) 2003-04-08 2005-05-19 Xingwu Wang Medical device with multiple coating layers
US20050079132A1 (en) 2003-04-08 2005-04-14 Xingwu Wang Medical device with low magnetic susceptibility
US20050025797A1 (en) 2003-04-08 2005-02-03 Xingwu Wang Medical device with low magnetic susceptibility
US20040254419A1 (en) 2003-04-08 2004-12-16 Xingwu Wang Therapeutic assembly
US7579077B2 (en) 2003-05-05 2009-08-25 Nanosys, Inc. Nanofiber surfaces for use in enhanced surface area applications
EP1618223A2 (en) 2003-04-28 2006-01-25 Nanosys, Inc. Super-hydrophobic surfaces, methods of their construction and uses therefor
TWI427709B (en) 2003-05-05 2014-02-21 Nanosys Inc Nanofiber surfaces for use in enhanced surface area applications
EP1636351A4 (en) 2003-05-21 2007-04-04 Gen Hospital Corp Microfabricated compositions and processes for engineering tissues containing multiple cell types
GB0313569D0 (en) 2003-06-12 2003-07-16 Plasso Technology Ltd Method
US7544770B2 (en) 2003-08-29 2009-06-09 Louisiana Tech Foundation, Inc. Multilayer films, coatings, and microcapsules comprising polypeptides
US7300801B2 (en) 2003-09-12 2007-11-27 3M Innovative Properties Company Welded sample preparation articles and methods
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US8592219B2 (en) 2005-01-17 2013-11-26 Gyros Patent Ab Protecting agent
US8158728B2 (en) 2004-02-13 2012-04-17 The University Of North Carolina At Chapel Hill Methods and materials for fabricating microfluidic devices
US7117807B2 (en) 2004-02-17 2006-10-10 University Of Florida Research Foundation, Inc. Dynamically modifiable polymer coatings and devices
US9016221B2 (en) 2004-02-17 2015-04-28 University Of Florida Research Foundation, Inc. Surface topographies for non-toxic bioadhesion control
US20050181353A1 (en) 2004-02-17 2005-08-18 Rao Galla C. Stabilization of cells and biological specimens for analysis
US20060057180A1 (en) 2004-02-20 2006-03-16 Ashutosh Chilkoti Tunable nonfouling surface of oligoethylene glycol
SE0400662D0 (en) 2004-03-24 2004-03-24 Aamic Ab Assay device and method
US20050215764A1 (en) 2004-03-24 2005-09-29 Tuszynski Jack A Biological polymer with differently charged portions
US7790458B2 (en) 2004-05-14 2010-09-07 Becton, Dickinson And Company Material and methods for the growth of hematopoietic stem cells
EP1744795A1 (en) 2004-05-14 2007-01-24 Becton, Dickinson and Company Articles having bioactive surfaces and solvent-free methods of preparation thereof
EP1753805A4 (en) 2004-05-17 2009-11-25 Univ Mcmaster Biological molecule-reactive hydrophilic silicone surface
JP2008501037A (en) 2004-06-01 2008-01-17 マイクロチップス・インコーポレーテッド Devices and methods for measuring and enhancing transport of drugs or analytes to / from medical implants
US7695775B2 (en) 2004-06-04 2010-04-13 Applied Microstructures, Inc. Controlled vapor deposition of biocompatible coatings over surface-treated substrates
US20060251795A1 (en) 2005-05-05 2006-11-09 Boris Kobrin Controlled vapor deposition of biocompatible coatings for medical devices
GB0420062D0 (en) 2004-09-09 2004-10-13 Akubio Ltd Assay methods,materials and preparations
US8663625B2 (en) 2004-10-15 2014-03-04 Cornell Research Foundation Diffusively permeable monolithic biomaterial with embedded microfluidic channels
WO2006050340A2 (en) 2004-11-01 2006-05-11 Polytechnic University Methods and apparatus for modifying gel adhesion strength
WO2006071640A2 (en) 2004-12-16 2006-07-06 The Regents Of The University Of Colorado Photolytic polymer surface modification
US8069782B2 (en) 2004-12-20 2011-12-06 Nanoink, Inc. Stamps with micrometer- and nanometer-scale features and methods of fabrication thereof
SE0403139D0 (en) 2004-12-23 2004-12-23 Nanoxis Ab Device and use thereof
US8158410B2 (en) 2005-01-18 2012-04-17 Biocept, Inc. Recovery of rare cells using a microchannel apparatus with patterned posts
US20090136982A1 (en) 2005-01-18 2009-05-28 Biocept, Inc. Cell separation using microchannel having patterned posts
EP1846567A4 (en) 2005-01-20 2009-12-09 Univ California Cellular microarrays for screening differentiation factors
US20090105463A1 (en) 2005-03-29 2009-04-23 Massachusetts Institute Of Technology Compositions of and Methods of Using Oversulfated Glycosaminoglycans
US20070196820A1 (en) 2005-04-05 2007-08-23 Ravi Kapur Devices and methods for enrichment and alteration of cells and other particles
GB2472927B (en) 2005-04-05 2011-05-04 Gen Hospital Corp Microfluidic Cell Capture on Micro-Corrugated Surface
US20070026417A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
EP1874920A4 (en) 2005-04-05 2009-11-04 Cellpoint Diagnostics Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US7485343B1 (en) 2005-04-13 2009-02-03 Sandia Corporation Preparation of hydrophobic coatings
US20060237390A1 (en) 2005-04-14 2006-10-26 King William P Combined Microscale Mechanical Topography and Chemical Patterns on Polymer Substrates for Cell Culture
US7846393B2 (en) 2005-04-21 2010-12-07 California Institute Of Technology Membrane filter for capturing circulating tumor cells
US7846743B2 (en) 2005-04-21 2010-12-07 California Institute Of Technology Uses of parylene membrane filters
CN100440572C (en) * 2005-05-31 2008-12-03 华南理工大学 Organic / macromolecule top emission light-emitting device and its application
US20070037173A1 (en) 2005-08-12 2007-02-15 Allard Jeffrey W Circulating tumor cells (CTC's): early assessment of time to progression, survival and response to therapy in metastatic cancer patients
SE528233C2 (en) 2005-06-20 2006-09-26 Aamic Ab Fluid handling device for handling fluid to be assayed, comprises absorbing zone(s) in fluid contact with second end of first passage and comprising projections perpendicular to substrate surface and capable of generating capillary flow
EP2004695A2 (en) 2005-07-08 2008-12-24 Xencor, Inc. Optimized anti-ep-cam antibodies
US7521195B1 (en) 2005-07-21 2009-04-21 Celera Corporation Lung disease targets and uses thereof
US8088161B2 (en) 2005-07-28 2012-01-03 Visioncare Ophthalmic Technologies Inc. Compressed haptics
US20070026416A1 (en) 2005-07-29 2007-02-01 Martin Fuchs Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US8921102B2 (en) 2005-07-29 2014-12-30 Gpb Scientific, Llc Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US7431969B2 (en) 2005-08-05 2008-10-07 Massachusetts Institute Of Technology Chemical vapor deposition of hydrogel films
US7993821B2 (en) 2005-08-11 2011-08-09 University Of Washington Methods and apparatus for the isolation and enrichment of circulating tumor cells
US7901950B2 (en) 2005-08-12 2011-03-08 Veridex, Llc Method for assessing disease states by profile analysis of isolated circulating endothelial cells
JP4941845B2 (en) 2005-08-24 2012-05-30 イーティーエイチ・チューリッヒ Method for producing catechol-equivalent molecules useful for surface modification
US20110097277A1 (en) 2005-08-25 2011-04-28 University Of Washington Particles coated with zwitterionic polymers
EP1924654B1 (en) 2005-08-25 2020-05-06 University of Washington Super-low fouling sulfobetaine and carboxybetaine materials and related methods
US20070048859A1 (en) 2005-08-25 2007-03-01 Sunsource Industries Closed system bioreactor apparatus
US20070059716A1 (en) 2005-09-15 2007-03-15 Ulysses Balis Methods for detecting fetal abnormality
US7713689B2 (en) 2005-09-15 2010-05-11 Duke University Non-fouling polymeric surface modification and signal amplification method for biomolecular detection
EP1946114A4 (en) 2005-09-21 2010-05-26 Ccc Diagnostics Llc Comprehensive diagnostic testing procedures for personalized anticancer chemotherapy (pac)
US7846445B2 (en) 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
JP4860703B2 (en) 2005-10-06 2012-01-25 ゼンコー・インコーポレイテッド Optimized anti-CD30 antibody
ES2341886T3 (en) * 2005-10-28 2010-06-29 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. IN VITRO FREE TRANSCRIPTION AND TRANSLATION OF MEMBRANE PROTEIN CELLS IN UNITED PLANE LIPID LAYERS.
EP1945754B1 (en) 2005-10-31 2014-07-23 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
WO2007056561A2 (en) 2005-11-09 2007-05-18 Liquidia Technologies, Inc. Medical device, materials, and methods
KR100763907B1 (en) * 2005-12-26 2007-10-05 삼성전자주식회사 A method of fabricating a microfluidic device and a microfluidic device fabricated by the same
WO2007079250A2 (en) 2005-12-29 2007-07-12 Cellpoint Diagnostics, Inc. Devices and methods for enrichment and alteration of circulating tumor cells and other particles
WO2007089911A2 (en) 2006-01-30 2007-08-09 The Scripps Research Institute Methods for detection of circulating tumor cells and methods of diagnosis of cancer in a mammalian subject
WO2007089762A2 (en) 2006-01-31 2007-08-09 Biomed Solutions, Llc Devices for selective recruitment, isolation, activation, and/or elimination of various cell populations
EP1984462B1 (en) 2006-02-10 2012-07-04 Université de Liège Electrografting method for forming and regulating a strong adherent nanostructured polymer coating
GB2448858A (en) 2006-02-15 2008-11-05 Aida Eng Ltd Microchannel chip and method for manufacturing such chip
DE102006009004A1 (en) 2006-02-23 2007-09-06 Sustech Gmbh & Co. Kg Multifunctional star-shaped prepolymers, their preparation and use
US20080213853A1 (en) 2006-02-27 2008-09-04 Antonio Garcia Magnetofluidics
BRPI0709600A2 (en) 2006-03-15 2011-07-19 Gen Hospital Corp Devices and methods for detecting cells and other analytes
WO2007121465A2 (en) 2006-04-18 2007-10-25 Wellstat Biologics Corporation Detection of proteins from circulating neoplastic cells
US8212083B2 (en) 2006-04-27 2012-07-03 Intezyne Technologies, Inc. Heterobifunctional poly(ethylene glycol) containing acid-labile amino protecting groups and uses thereof
CA2861601A1 (en) 2006-04-27 2007-11-08 Intezyne Technologies, Inc. Poly (ethylene glycol) containing chemically disparate endgroups
US20090311734A1 (en) 2006-05-12 2009-12-17 Jan Greve Laser Illumination System in Fluorescent Microscopy
ATE542583T1 (en) 2006-05-22 2012-02-15 Univ Columbia METHOD FOR MEMBRANE-LESS MICROFLUID EXCHANGE IN AN H-FILTER AND FILTERING OF THE EXTRACTION FLUID OUTPUT STREAMS
WO2008016414A2 (en) 2006-06-01 2008-02-07 The Trustees Of Princeton University Apparatus and method for continuous particle separation
WO2007140595A1 (en) 2006-06-06 2007-12-13 Societe De Commercialisation Des Produits De La Recherche Appliquee - Socpra-Sciences Et Genie S.E.C. Assay supports comprising a peg support, said support attached from a peg solution in cloud point (theta solvent) conditions
WO2007147018A1 (en) 2006-06-14 2007-12-21 Cellpoint Diagnostics, Inc. Analysis of rare cell-enriched samples
US8535791B2 (en) 2006-06-30 2013-09-17 The University Of Akron Aligned carbon nanotube-polymer materials, systems and methods
CA2657621A1 (en) 2006-07-14 2008-01-17 Aviva Biosciences Corporation Methods and compositions for detecting rare cells from a biological sample
WO2008019381A1 (en) * 2006-08-07 2008-02-14 University Of Washington Mixed charge copolymers and hydrogels
CN101652126B (en) 2006-08-08 2013-07-17 得克萨斯大学体系董事会 Multistage delivery of active agents
US8114431B2 (en) 2006-09-19 2012-02-14 Georgia Tech Research Corporation Biomolecular coating for implants
GB0618460D0 (en) 2006-09-20 2006-11-01 Univ Belfast Process for preparing surfaces with tailored wettability
EP2078062B1 (en) 2006-10-19 2018-12-05 Northwestern University Surface-independent, surface-modifying, multifunctional coatings and applications thereof
CA2666378A1 (en) 2006-11-03 2008-05-08 The Governors Of The University Of Alberta Microfluidic device having an array of spots
US20080114096A1 (en) 2006-11-09 2008-05-15 Hydromer, Inc. Lubricious biopolymeric network compositions and methods of making same
US20080113350A1 (en) 2006-11-09 2008-05-15 Terstappen Leon W M M Blood test to monitor the genetic changes of progressive cancer using immunomagnetic enrichment and fluorescence in situ hybridization (FISH)
GB0623160D0 (en) 2006-11-20 2006-12-27 Smith & Nephew Biomolecules
WO2008064058A2 (en) 2006-11-21 2008-05-29 Abbott Laboratories Use of a terpolymer of tetrafluoroethylene, hexafluoropropylene, and vinylidene fluoride in drug eluting coatings
FR2909013B1 (en) 2006-11-28 2011-02-25 Commissariat Energie Atomique THIN FILM COATING PROCESS
WO2008083390A2 (en) 2006-12-29 2008-07-10 University Of Washington Dual-functional nonfouling surfaces and materials
WO2008089282A2 (en) 2007-01-16 2008-07-24 Silver James H Sensors for detecting subtances indicative of stroke, ischemia, infection or inflammation
WO2008108980A2 (en) 2007-03-01 2008-09-12 Nanospectra Biosciences, Inc. Devices and methods for extracorporeal ablation of circulating cells
US7981688B2 (en) 2007-03-08 2011-07-19 University Of Washington Stimuli-responsive magnetic nanoparticles and related methods
WO2008121375A2 (en) 2007-03-29 2008-10-09 Pacific Biosciences Of California, Inc. Modified surfaces for immobilization of active molecules
PT2142570E (en) 2007-04-04 2011-09-13 Sigma Tau Ind Farmaceuti Anti-epcam antibody and uses thereof
US20100233694A1 (en) 2007-04-16 2010-09-16 On-O-ity, Inc Devices and methods for diagnosing, prognosing, or theranosing a condition by enriching rare cells
WO2008130977A2 (en) 2007-04-16 2008-10-30 The General Hospital Corporation D/B/A Massachusetts General Hospital Systems and methods for particle focusing in microchannels
WO2008131301A1 (en) * 2007-04-18 2008-10-30 Massachusetts Institute Of Technology Surfaces, methods and devices employing cell rolling
US8945909B2 (en) 2007-04-25 2015-02-03 The Regents Of The University Of Michigan Tunable elastomeric nanochannels for nanofluidic manipulation
US10202711B2 (en) 2007-05-09 2019-02-12 Massachusetts Institute Of Technology Tunable surface
EP2149049A4 (en) 2007-05-18 2010-07-07 Fujirebio Kk Chemical surface nanopatterns to increase activity of surface-immobilized biomolecules
DK2176298T3 (en) 2007-05-30 2018-02-12 Xencor Inc Methods and compositions for inhibiting CD32B-expressing cells
US20080312356A1 (en) 2007-06-13 2008-12-18 Applied Mcrostructures, Inc. Vapor-deposited biocompatible coatings which adhere to various plastics and metal
US8841135B2 (en) 2007-06-20 2014-09-23 University Of Washington Biochip for high-throughput screening of circulating tumor cells
US8658093B2 (en) 2007-07-06 2014-02-25 Applied Biosystems, Llc Devices and methods for the detection of analytes
EP2020261A1 (en) 2007-07-20 2009-02-04 Nederlandse Organisatie voor toegepast- natuurwetenschappelijk onderzoek TNO Multi component particle generating system
DE102007039665A1 (en) 2007-08-22 2009-02-26 Sustech Gmbh & Co. Kg Silyl-functional linear prepolymers, their preparation and use
EP2037265A1 (en) 2007-09-17 2009-03-18 Adnagen AG Solid phase cell isolation and/or enrichment method
US7736891B2 (en) 2007-09-11 2010-06-15 University Of Washington Microfluidic assay system with dispersion monitoring
US11385226B2 (en) 2007-09-17 2022-07-12 Red Ivory Llc Self-actuating signal producing detection devices and methods
US9150788B2 (en) 2007-09-18 2015-10-06 Syracuse University Non-amphiphile-based water-in-water emulsion and uses thereof
WO2009079053A2 (en) 2007-09-19 2009-06-25 Massachusetts Institute Of Technology High affinity metal-oxide binding peptides with reversible binding
CN101918543A (en) 2007-09-27 2010-12-15 麻省理工学院 Cell rolling separates
US20090098017A1 (en) 2007-10-16 2009-04-16 Board Of Regents, The University Of Texas System Nanoporous membrane exchanger
WO2009051734A1 (en) 2007-10-17 2009-04-23 The General Hospital Corporation Microchip-based devices for capturing circulating tumor cells and methods of their use
US8992471B2 (en) 2007-11-05 2015-03-31 Nanocopoeia, Inc. Coated devices and method of making coated devices that reduce smooth muscle cell proliferation and platelet activity
CA2705485C (en) 2007-11-19 2016-03-08 University Of Washington Cationic betaine precursors to zwitterionic betaines having controlled biological properties
US8658192B2 (en) 2007-11-19 2014-02-25 University Of Washington Integrated antimicrobial and low fouling materials
US20090181441A1 (en) 2007-11-27 2009-07-16 Board Of Trustees Of Michigan State University Porous silicon-polymer composites for biosensor applications
EP2229441B1 (en) 2007-12-12 2014-10-01 The Board of Trustees of The Leland Stanford Junior University Method and apparatus for magnetic separation of cells
CA2709550A1 (en) 2007-12-17 2009-06-25 Lux Innovate Limited Compositions and methods for maintenance of fluid conducting and containment systems
WO2009079664A1 (en) 2007-12-19 2009-06-25 Georgia Tech Research Corporation Modification of biomaterials with microgel films
EP2980217A1 (en) * 2007-12-31 2016-02-03 XOMA Technology Ltd. Methods and materials for targeted mutagenesis
GB0801600D0 (en) 2008-01-29 2008-03-05 Univ Cardiff Microtrench
JP2011514182A (en) 2008-02-04 2011-05-06 ザ トラスティーズ オブ コロンビア ユニバーシティ イン ザ シティ オブ ニューヨーク Fluid separation apparatus, system, and method
US20100323918A1 (en) 2008-02-10 2010-12-23 Microdysis, Inc Polymer surface functionalization and related applications
US8367370B2 (en) 2008-02-11 2013-02-05 Wheeler Aaron R Droplet-based cell culture and cell assays using digital microfluidics
US8008032B2 (en) 2008-02-25 2011-08-30 Cellective Dx Corporation Tagged ligands for enrichment of rare analytes from a mixed sample
WO2009111159A2 (en) 2008-03-03 2009-09-11 New York University Biocompatible materials containing stable complexes of tsg-6 and hyaluronan and method of using same
WO2009120151A1 (en) 2008-03-28 2009-10-01 Nanyang Technological University Membrane made of a nanostructured material
US8796184B2 (en) 2008-03-28 2014-08-05 Sentilus, Inc. Detection assay devices and methods of making and using the same
US8128983B2 (en) 2008-04-11 2012-03-06 Abbott Cardiovascular Systems Inc. Coating comprising poly(ethylene glycol)-poly(lactide-glycolide-caprolactone) interpenetrating network
US8177978B2 (en) 2008-04-15 2012-05-15 Nanoh20, Inc. Reverse osmosis membranes
US8567612B2 (en) 2008-04-15 2013-10-29 Nanoh2O, Inc. Hybrid TFC RO membranes with nitrogen additives
SE533515C2 (en) 2008-04-16 2010-10-12 Aamic Ab Analysis procedure for simultaneous analysis of several analytes
US8916188B2 (en) 2008-04-18 2014-12-23 Abbott Cardiovascular Systems Inc. Block copolymer comprising at least one polyester block and a poly (ethylene glycol) block
US20090264317A1 (en) 2008-04-18 2009-10-22 University Of Massachusetts Functionalized nanostructure, methods of manufacture thereof and articles comprising the same
US20090285873A1 (en) 2008-04-18 2009-11-19 Abbott Cardiovascular Systems Inc. Implantable medical devices and coatings therefor comprising block copolymers of poly(ethylene glycol) and a poly(lactide-glycolide)
JP5042110B2 (en) 2008-04-22 2012-10-03 サルナス、ペトロニス Production of nanopores
WO2009140326A2 (en) 2008-05-16 2009-11-19 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Microfluidic isolation of tumor cells or other rare cells from whole blood or other liquids
GB0810039D0 (en) 2008-06-03 2008-07-09 Univ Belfast Shape-formed product with tailored wettability
EP2300207A4 (en) 2008-06-26 2012-05-09 Harvard College Versatile high aspect ratio actuatable nanostructured materials through replication
WO2010123608A2 (en) * 2009-01-29 2010-10-28 The Regents Of The University Of California A spatial biomarker of disease and detection of spatial organization of cellular recptors
EP3196642A1 (en) 2008-07-24 2017-07-26 The Trustees of Princeton University Bump array device having asymmetric gaps for segregation of particles
WO2010017671A1 (en) 2008-08-11 2010-02-18 Peking University Superhydrophobic poly(dimethylsiloxane) and methods for making the same
US20100055733A1 (en) 2008-09-04 2010-03-04 Lutolf Matthias P Manufacture and uses of reactive microcontact printing of biomolecules on soft hydrogels
WO2010028160A1 (en) 2008-09-05 2010-03-11 The Scripps Research Institute Methods for the detection of circulating tumor cells
US8367090B2 (en) 2008-09-05 2013-02-05 Abbott Cardiovascular Systems Inc. Coating on a balloon comprising a polymer and a drug
US20100096327A1 (en) 2008-09-19 2010-04-22 Gin Douglas L Polymer coatings that resist adsorption of proteins
US10314524B2 (en) 2008-09-23 2019-06-11 Gilupi Gmbh Diagnostic analyte collection device based on flexible polymers with biological surface modification and microfluidic functionality
US20110250679A1 (en) 2008-10-02 2011-10-13 The Regents Of The University Of California Methods and Compositions for High-Resolution Micropatterning for Cell Culture
WO2010042943A1 (en) 2008-10-10 2010-04-15 Massachusetts Institute Of Technology Tunable hydrogel microparticles
WO2010041231A2 (en) 2008-10-10 2010-04-15 Cnrs-Dae Cell sorting device
WO2010053993A1 (en) 2008-11-04 2010-05-14 The Trustees Of Columbia University In The City Of New York Heterobifunctional polymers and methods for layer-by-layer construction of multilayer films
US8292492B2 (en) 2008-11-11 2012-10-23 Sandia Corporation Airfoil-shaped micro-mixers for reducing fouling on membrane surfaces
ES2432388T3 (en) 2008-12-05 2013-12-03 Semprus Biosciences Corporation Anti-fouling, antimicrobial and antithrombogenic graft compositions from the surface
US8308699B2 (en) 2008-12-05 2012-11-13 Semprus Biosciences Corp. Layered non-fouling, antimicrobial antithrombogenic coatings
EP2373715A4 (en) 2008-12-08 2014-12-03 Univ Washington Omega-functionalized polymers, junction-functionalized block copolymers, polymer bioconjugates, and radical chain extension polymerization
JP2012519558A (en) 2009-03-10 2012-08-30 モナッシュ ユニヴァーシティ Platelet aggregation using microfluidic devices
US9140697B2 (en) 2009-03-18 2015-09-22 The Regents Of The University Of California Device for capturing circulating cells
KR101796906B1 (en) 2009-03-24 2017-11-10 유니버시티 오브 시카고 Method for carrying out a reaction
US8343440B2 (en) 2009-03-27 2013-01-01 Seiko Epson Corporation Cell separating apparatus and cell separating method
US8569463B2 (en) 2009-04-23 2013-10-29 Syracuse University Method of covalently modifying proteins with organic molecules to prevent aggregation
EP2421628A4 (en) 2009-04-23 2012-10-10 Logos Energy Inc Lateral displacement array for microfiltration
US20120077246A1 (en) 2009-04-24 2012-03-29 The Board Of Trustees Of The University Of Illinoi Methods and Devices for Capturing Circulating Tumor Cells
WO2010132795A2 (en) 2009-05-15 2010-11-18 The General Hospital Corporation Systems, devices, and methods for specific capture and release of biological sample components
CN102803504B (en) 2009-06-15 2015-07-01 3M创新有限公司 Detection of acid-producing bacteria
AU2010289448A1 (en) 2009-09-03 2012-03-22 The Scripps Research Institute Method for categorizing circulating tumor cells
US8481336B2 (en) 2009-09-09 2013-07-09 The Board Of Trustees Of The Leland Stanford Junior University Magnetic separation device for cell sorting and analysis
JP6048660B2 (en) 2009-09-21 2016-12-21 ランジュ ラルハン Methods and compositions for diagnosing and treating thyroid cancer
WO2011038138A1 (en) 2009-09-25 2011-03-31 Origene Technologies, Inc. Protein arrays and uses thereof
CN101701039B (en) * 2009-11-06 2011-10-26 中国人民解放军第四军医大学 Variable regions of light chains and heavy chains of FMU-EPCAM-2A9 monoclonal antibodies
US20110165161A1 (en) 2009-12-23 2011-07-07 Shih-Yao Lin Anti-epcam antibodies that induce apoptosis of cancer cells and methods using same
US20110224383A1 (en) 2010-03-11 2011-09-15 Intezyne Technologies, Inc. Poly(ethylene glycol) derivatives for metal-free click chemistry
SG185100A1 (en) 2010-05-04 2012-11-29 Paul Walfish Method for the diagnosis of epithelial cancers by the detection of epicd polypeptide
TWI539158B (en) 2010-06-08 2016-06-21 維里德克斯有限責任公司 A method of predicting clinical outcomes for melanoma patients using circulating melanoma cells in blood
WO2011156612A2 (en) 2010-06-09 2011-12-15 Semprus Biosciences Corp. Articles having non-fouling surfaces and processes for preparing the same including applying a primer coat
US20110305898A1 (en) 2010-06-09 2011-12-15 Zheng Zhang Non-fouling, anti-microbial, anti-thrombogenic graft compositions
US9096703B2 (en) 2010-06-09 2015-08-04 Semprus Biosciences Corporation Non-fouling, anti-microbial, anti-thrombogenic graft-from compositions
US9248467B2 (en) 2010-07-13 2016-02-02 Rigoberto Advincula Types of electrodeposited polymer coatings with reversible wettability and electro-optical properties
WO2012016136A2 (en) 2010-07-30 2012-02-02 The General Hospital Corporation Microscale and nanoscale structures for manipulating particles
EP2603793A4 (en) 2010-08-15 2014-03-19 Gpb Scientific Llc Microfluidic cell separation in the assay of blood
US20120058302A1 (en) 2010-09-03 2012-03-08 Massachusetts Institute Of Technology Fabrication of anti-fouling surfaces comprising a micro- or nano-patterned coating
CN104741157B (en) 2010-09-14 2017-04-12 加利福尼亚大学董事会 Device for isolating cells from heterogeneous solution using microfluidic trapping vortices
CN102011193B (en) * 2010-09-21 2013-03-13 南京航空航天大学 Protein modified GaN nanowire array as well as preparation method and application thereof
CN103889556A (en) 2011-01-06 2014-06-25 Gpb科学有限责任公司 Circulating tumor cell capture on a microfluidic chip incorporating both affinity and size
US20140308669A1 (en) 2011-01-24 2014-10-16 Epic Science, Inc. Methods for obtaining single cells and applications of single cell omics
EP2678042B1 (en) 2011-02-23 2018-05-09 The Board of Trustees of the University of Illionis Amphiphilic dendron-coils, micelles thereof and uses
USD650091S1 (en) 2011-04-19 2011-12-06 Zach Odeh Microfluidic device
KR101279918B1 (en) 2011-05-27 2013-07-01 한국과학기술연구원 Device for detection of tumor cells and detecting method tumor cells
US9541480B2 (en) 2011-06-29 2017-01-10 Academia Sinica Capture, purification, and release of biological substances using a surface coating
EP2729802B1 (en) 2011-07-07 2015-05-06 Scripps Health Method of analyzing cardiovascular disorders and uses thereof
WO2013036620A1 (en) 2011-09-06 2013-03-14 Becton, Dickinson And Company Methods and compositions for cytometric detection of rare target cells in a sample
US20140296095A1 (en) 2011-09-23 2014-10-02 The Trustees Of Columbia University In The City Of New York Spatially Selective Release of Aptamer-Captured Cells by Temperature Mediation
JP6163502B2 (en) 2012-03-02 2017-07-12 アカデミア シニカAcademia Sinica Anti-epithelial cell adhesion molecule (EpCAM) antibody and method of use thereof
US9494500B2 (en) 2012-10-29 2016-11-15 Academia Sinica Collection and concentration system for biologic substance of interest and use thereof
CN106662514A (en) 2014-04-01 2017-05-10 中央研究院 Methods and systems for cancer diagnosis and prognosis
EP2998026B1 (en) 2014-08-26 2024-01-17 Academia Sinica Collector architecture layout design
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11674958B2 (en) 2011-06-29 2023-06-13 Academia Sinica Capture, purification, and release of biological substances using a surface coating
US10495644B2 (en) 2014-04-01 2019-12-03 Academia Sinica Methods and systems for cancer diagnosis and prognosis
US10112198B2 (en) 2014-08-26 2018-10-30 Academia Sinica Collector architecture layout design
US10107726B2 (en) 2016-03-16 2018-10-23 Cellmax, Ltd. Collection of suspended cells using a transferable membrane
US10605708B2 (en) 2016-03-16 2020-03-31 Cellmax, Ltd Collection of suspended cells using a transferable membrane
WO2019025437A1 (en) 2017-07-31 2019-02-07 Universidad De Cantabria Cartridge, device and method for detecting, capturing, identifying and counting circulating tumour cells

Also Published As

Publication number Publication date
EP2726870B1 (en) 2018-10-03
EP2726870A2 (en) 2014-05-07
CN107315086B (en) 2019-09-10
US20210088514A1 (en) 2021-03-25
CN107315086A (en) 2017-11-03
TWI550273B (en) 2016-09-21
US9541480B2 (en) 2017-01-10
TW201323875A (en) 2013-06-16
US11674958B2 (en) 2023-06-13
CN110763842A (en) 2020-02-07
US20140255976A1 (en) 2014-09-11
EP2726870A4 (en) 2015-03-11
WO2013003624A3 (en) 2013-03-14
CN103998932A (en) 2014-08-20
TWI599765B (en) 2017-09-21
US20230366879A1 (en) 2023-11-16
TW201636593A (en) 2016-10-16
CN103998932B (en) 2017-06-06
WO2013003624A2 (en) 2013-01-03

Similar Documents

Publication Publication Date Title
US11674958B2 (en) Capture, purification, and release of biological substances using a surface coating
US9494500B2 (en) Collection and concentration system for biologic substance of interest and use thereof
US10011817B2 (en) Cell rolling separation
Ishihara et al. Bioinspired interface for nanobiodevices based on phospholipid polymer chemistry
US10900969B2 (en) Biomimetic microfluid device for capturing circulating tumor cells
US20200009225A1 (en) Method to functionalize cells in human blood, other fluids and tissues using nanoparticles
TWI638989B (en) A microfluidic chip for selectively enriching rare cell
JP7378212B2 (en) Composition for cell adhesion and base material for cell adhesion
TWI660175B (en) A surface coating for the capture of a circulating tumor cell
JP6400483B2 (en) Nano gap structure type substrate
JP2002537025A (en) Biocompatible materials with new functions
Choudhury Cross-flow Microfiltration for Isolation, Selective Capture, and Release of Liposarcoma Extracellular Vesicles
Dong Layer-by-layer assembly of responsive polymeric nanofilms: Characterizations and new applications

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION